351
|
Abstract
Adhesion G protein-coupled receptors (aGPCRs) have a long evolutionary history dating back to very basal unicellular eukaryotes. Almost every vertebrate is equipped with a set of different aGPCRs. Genomic sequence data of several hundred extinct and extant species allows for reconstruction of aGPCR phylogeny in vertebrates and non-vertebrates in general but also provides a detailed view into the recent evolutionary history of human aGPCRs. Mining these sequence sources with bioinformatic tools can unveil many facets of formerly unappreciated aGPCR functions. In this review, we extracted such information from the literature and open public sources and provide insights into the history of aGPCR in humans. This includes comprehensive analyses of signatures of selection, variability of human aGPCR genes, and quantitative traits at human aGPCR loci. As indicated by a large number of genome-wide genotype-phenotype association studies, variations in aGPCR contribute to specific human phenotypes. Our survey demonstrates that aGPCRs are significantly involved in adaptation processes, phenotype variations, and diseases in humans.
Collapse
Affiliation(s)
- Peter Kovacs
- Integrated Research and Treatment Center (IFB) AdiposityDiseases, Medical Faculty, University of Leipzig, Liebigstr. 21, Leipzig, 04103, Germany.
| | - Torsten Schöneberg
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, Leipzig, 04103, Germany.
| |
Collapse
|
352
|
Sandholt CH, Grarup N, Pedersen O, Hansen T. Genome-wide association studies of human adiposity: Zooming in on synapses. Mol Cell Endocrinol 2015; 418 Pt 2:90-100. [PMID: 26427653 DOI: 10.1016/j.mce.2015.09.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 02/03/2023]
Abstract
The decade anniversary for genome-wide association studies (GWAS) is approaching, and this experimental approach has commenced a deeper understanding of the genetics underlying complex diseases. In obesity genetics the GIANT (Genetic Investigation of ANthropometric Traits) consortium has played a crucial role, recently with two comprehensive meta-analyses, one focusing on general obesity, analyzing body-mass index (BMI) and the other on fat distribution, focusing on waist-hip ratio adjusted for BMI. With the in silico methods applied in these two studies as the pivot, this review looks into some of the biological knowledge, beginning to emerge from the intricate genomic background behind the genetic determinants of human adiposity. These include synaptic dysfunction, where GWAS pinpoint potential new mechanisms in pathways already known to be linked with obesity.
Collapse
Affiliation(s)
- Camilla H Sandholt
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
353
|
Hunt LE, Noyvert B, Bhaw-Rosun L, Sesay AK, Paternoster L, Nohr EA, Davey Smith G, Tommerup N, Sørensen TIA, Elgar G. Complete re-sequencing of a 2Mb topological domain encompassing the FTO/IRXB genes identifies a novel obesity-associated region upstream of IRX5. Genome Med 2015; 7:126. [PMID: 26642925 PMCID: PMC4671217 DOI: 10.1186/s13073-015-0250-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/17/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Association studies have identified a number of loci that contribute to an increased body mass index (BMI), the strongest of which is in the first intron of the FTO gene on human chromosome 16q12.2. However, this region is both non-coding and under strong linkage disequilibrium, making it recalcitrant to functional interpretation. Furthermore, the FTO gene is located within a complex cis-regulatory landscape defined by a topologically associated domain that includes the IRXB gene cluster, a trio of developmental regulators. Consequently, at least three genes in this interval have been implicated in the aetiology of obesity. METHODS Here, we sequence a 2 Mb region encompassing the FTO, RPGRIP1L and IRXB cluster genes in 284 individuals from a well-characterised study group of Danish men containing extremely overweight young adults and controls. We further replicate our findings both in an expanded male cohort and an independent female study group. Finally, we compare our variant data with a previous study describing IRX3 and FTO interactions in this region. RESULTS We obtain deep coverage across the entire region, allowing accurate and unequivocal determination of almost every single nucleotide polymorphism and short insertion/deletion. As well as confirming previous findings across the interval, we identify a further novel age-dependent association upstream of IRX5 that imposes a similar burden on BMI to the FTO locus. CONCLUSIONS Our findings are consistent with the hypothesis that chromatin architectures play a role in regulating gene expression levels across topological domains while our targeted sequence approach represents a widely applicable methodology for high-resolution analysis of regional variation across candidate genomic loci.
Collapse
Affiliation(s)
- Lilian E Hunt
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Boris Noyvert
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Leena Bhaw-Rosun
- Genomics Facility, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Abdul K Sesay
- Genomics Facility, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, University of Bristol, Bristol, UK
| | - Ellen A Nohr
- Research Unit for Gynecology and Obstetrics, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, University of Bristol, Bristol, UK
| | - Niels Tommerup
- Willhelm Johannsen Centre for Functional Genome Research, Department of Cellular and Molecular Medicine, The Faculty of Health Sciences, The University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark
| | - Thorkild I A Sørensen
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, University of Bristol, Bristol, UK.,The Novo Nordisk Foundation Centre for Basic Metabolic Research, Section on Metabolic genetics, The Faculty of Health and Medical Sciences, University of Copenhagen, DK2100, Copenhagen Ø, Denmark.,Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospitals, The Capital Region, DK2000, Frederiksberg, Denmark
| | - Greg Elgar
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK.
| |
Collapse
|
354
|
Artigas MS, Wain LV, Miller S, Kheirallah AK, Huffman JE, Ntalla I, Shrine N, Obeidat M, Trochet H, McArdle WL, Alves AC, Hui J, Zhao JH, Joshi PK, Teumer A, Albrecht E, Imboden M, Rawal R, Lopez LM, Marten J, Enroth S, Surakka I, Polasek O, Lyytikäinen LP, Granell R, Hysi PG, Flexeder C, Mahajan A, Beilby J, Bossé Y, Brandsma CA, Campbell H, Gieger C, Gläser S, González JR, Grallert H, Hammond CJ, Harris SE, Hartikainen AL, Heliövaara M, Henderson J, Hocking L, Horikoshi M, Hutri-Kähönen N, Ingelsson E, Johansson Å, Kemp JP, Kolcic I, Kumar A, Lind L, Melén E, Musk AW, Navarro P, Nickle DC, Padmanabhan S, Raitakari OT, Ried JS, Ripatti S, Schulz H, Scott RA, Sin DD, Starr JM, Viñuela A, Völzke H, Wild SH, Wright AF, Zemunik T, Jarvis DL, Spector TD, Evans DM, Lehtimäki T, Vitart V, Kähönen M, Gyllensten U, Rudan I, Deary IJ, Karrasch S, Probst-Hensch NM, Heinrich J, Stubbe B, Wilson JF, Wareham NJ, James AL, Morris AP, Jarvelin MR, Hayward C, Sayers I, Strachan DP, Hall IP, Tobin MD. Sixteen new lung function signals identified through 1000 Genomes Project reference panel imputation. Nat Commun 2015; 6:8658. [PMID: 26635082 PMCID: PMC4686825 DOI: 10.1038/ncomms9658] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 09/17/2015] [Indexed: 01/11/2023] Open
Abstract
Lung function measures are used in the diagnosis of chronic obstructive pulmonary disease. In 38,199 European ancestry individuals, we studied genome-wide association of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and FEV1/FVC with 1000 Genomes Project (phase 1)-imputed genotypes and followed up top associations in 54,550 Europeans. We identify 14 novel loci (P<5 × 10(-8)) in or near ENSA, RNU5F-1, KCNS3, AK097794, ASTN2, LHX3, CCDC91, TBX3, TRIP11, RIN3, TEKT5, LTBP4, MN1 and AP1S2, and two novel signals at known loci NPNT and GPR126, providing a basis for new understanding of the genetic determinants of these traits and pulmonary diseases in which they are altered.
Collapse
Affiliation(s)
- María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Louise V. Wain
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Suzanne Miller
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Abdul Kader Kheirallah
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Ioanna Ntalla
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Nick Shrine
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Ma'en Obeidat
- University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, British Columbia, Canada V6Z 1Y6
| | - Holly Trochet
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, MRC -PHE Centre for Environment & Health, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Jennie Hui
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- PathWest Laboratory Medicine WA, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
- School of Population Health, The University of Western Australia, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, The University of Western Australia, Western Australia 6009, Australia
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0SL, UK
| | - Peter K. Joshi
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Alexander Teumer
- University Medicine Greifswald, Community Medicine, SHIP—Clinical Epidemiological Research, Greifswald 17489, Germany
- Department for Genetics and Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald 17489, Germany
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
| | - Rajesh Rawal
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Lorna M. Lopez
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Department of Psychology, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Jonathan Marten
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Stefan Enroth
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala 751 23, Sweden
| | - Ida Surakka
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FI-00014, Finland
- The National Institute for Health and Welfare (THL), Helsinki FI-00271, Finland
| | - Ozren Polasek
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
- Department of Public Health, Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere FI-33520, Finland
| | - Raquel Granell
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Pirro G. Hysi
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Claudia Flexeder
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - John Beilby
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- PathWest Laboratory Medicine WA, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, The University of Western Australia, Western Australia 6009, Australia
| | - Yohan Bossé
- Department of Molecular Medicine, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec, Canada G1V 0A6
| | - Corry-Anke Brandsma
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9700, The Netherlands
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Sven Gläser
- Department of Internal Medicine B, Pneumology, Cardiology, Intensive Care, Weaning, Field of Research: Pneumological Epidemiology, University Medicine Greifswald, Greifswald 17489, Germany
| | - Juan R. González
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona E-08003, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain
- Pompeu Fabra University (UPF), Barcelona 08002, Catalonia, Spain
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Chris J. Hammond
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Sarah E. Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Anna-Liisa Hartikainen
- Department of Obstetrics and Gynecology of Oulu University Hospital ,MRC of Oulu University, Oulu 90220, Finland
| | - Markku Heliövaara
- The National Institute for Health and Welfare (THL), Helsinki FI-00271, Finland
| | - John Henderson
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Lynne Hocking
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
- Division of Applied Health Sciences, University of Aberdeen, Aberdeen, Scotland AB24 3FX, UK
| | - Momoko Horikoshi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX1 2JD, UK
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, Tampere 33521, Finland
- Department of Pediatrics, University of Tampere School of Medicine, Tampere FI-33520, Finland
| | - Erik Ingelsson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala 751 23, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Åsa Johansson
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala 751 23, Sweden
- Uppsala Clinical Research Centre, Uppsala University, Uppsala 751 23, Sweden
| | - John P. Kemp
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland QLD 4072, Australia
- MRC Integrative Epidemiology Unit, Bristol BS8 1TH, UK
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Ashish Kumar
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 7, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala 751 23, Sweden
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Sachs' Children's Hospital, Stockholm SE-171 7, Sweden
| | - Arthur W. Musk
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Western Australia 6009, Australia
| | - Pau Navarro
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - David C. Nickle
- Genetics and Pharmacogenomics, Merck Research Labs, Boston, Massachusetts 02115, USA
| | - Sandosh Padmanabhan
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
- Division of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, Scotland, UK
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20520, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20014, Finland
| | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FI-00014, Finland
- Department of Public Health, University of Helsinki, Helsinki FI-00014, Finland
- Department of Human Genomics, Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Holger Schulz
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich 85764, Germany
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0SL, UK
| | - Don D. Sin
- University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, British Columbia, Canada V6Z 1Y6
- Respiratory Division, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z4
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Alzheimer Scotland Research Centre, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Ana Viñuela
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Henry Völzke
- University Medicine Greifswald, Community Medicine, SHIP—Clinical Epidemiological Research, Greifswald 17489, Germany
| | - Sarah H. Wild
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Alan F. Wright
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Tatijana Zemunik
- Department of Medical Biology, Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Deborah L. Jarvis
- Respiratory Epidemiology and Public Health, Imperial College London, London SW7 2AZ, UK
- MRC Health Protection Agency (HPA) Centre for Environment and Health, Imperial College London, London SW7 2AZ, UK
| | - Tim D. Spector
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - David M. Evans
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland QLD 4072, Australia
- MRC Integrative Epidemiology Unit, Bristol BS8 1TH, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere FI-33520, Finland
| | - Veronique Vitart
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere and Tampere University Hospital, Tampere 33521, Finland
| | - Ulf Gyllensten
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala 751 23, Sweden
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
- Centre for Population Health Sciences, Medical School, University of Edinburgh, Edinburgh EH8 9AD, Scotland, UK
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Department of Psychology, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Stefan Karrasch
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Institute of General Practice, University Hospital Klinikum rechts der Isar, Technische Universität München, Munich D - 81675, Germany
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Ludwig-Maximilians-Universität, Munich 80539, Germany
| | - Nicole M. Probst-Hensch
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich 85764, Germany
- University Hospital Munich, Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Ludwig-Maximilian University Munich, Munich 80539, Germany
| | - Beate Stubbe
- Department of Internal Medicine B, Pneumology, Cardiology, Intensive Care, Weaning, Field of Research: Pneumological Epidemiology, University Medicine Greifswald, Greifswald 17489, Germany
| | - James F. Wilson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0SL, UK
| | - Alan L. James
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Western Australia 6009, Australia
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 7ZX, UK
- Estonian Genome Centre, University of Tartu, Tartu 50090, Estonia
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC -PHE Centre for Environment & Health, School of Public Health, Imperial College London, London SW7 2AZ, UK
- Center for Life Course Epidemiology, Faculty of Medicine, P.O.Box 5000, FI-90014 University of Oulu, Oulu FI-01051, Finland
- Biocenter Oulu, P.O.Box 5000, Aapistie 5A, FI-90014 University of Oulu, Oulu FI-01051, Finland
- Unit of Primary Care, Oulu University Hospital, Kajaanintie 50, P.O.Box 20, FI-90220 Oulu, 90029 OYS, Finland
| | - Caroline Hayward
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
| | - Ian Sayers
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - David P. Strachan
- Population Health Research Institute, St George's, University of London, Cranmer Terrace, London WC1B 5DN, UK
| | - Ian P. Hall
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
355
|
Niesor EJ, Benghozi R, Amouyel P, Ferdinand KC, Schwartz GG. Adenylyl Cyclase 9 Polymorphisms Reveal Potential Link to HDL Function and Cardiovascular Events in Multiple Pathologies: Potential Implications in Sickle Cell Disease. Cardiovasc Drugs Ther 2015; 29:563-572. [PMID: 26619842 DOI: 10.1007/s10557-015-6626-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Adenylyl cyclase 9 (ADCY9) mediates β2-adrenoceptor (β2-AR) signalling. Both proteins are associated with caveolae, specialized cholesterol-rich membrane substructures. Apolipoprotein A1 (ApoA1), the major protein component of high-density lipoprotein (HDL), removes cholesterol from cell membrane and caveolae and may thereby influence β2-AR signalling, shown in vitro to be modulated by cholesterol. Patients with Sickle Cell Disease (SCD) typically have low HDL and ApoA1 levels. In patients, mainly of African origin, with SCD, β2-AR activation may trigger adhesion of red blood cells to endothelial cells, leading to vascular occlusive events. Moreover, ADCY9 polymorphism is associated with risk of stroke in SCD. In recent clinical trials, ADCY9 polymorphism was found to be a discriminant factor associated with the risk of cardiovascular (CV) events in Caucasian patients treated with the HDL-raising compound dalcetrapib. We hypothesize that these seemingly disparate observations share a common mechanism related to interaction of HDL/ApoA1 and ADCY9 on β2-AR signalling. This review also raises the importance of characterizing polymorphisms that determine the response to HDL-raising and -mimicking agents in the non-Caucasian population at high risk of CV diseases and suffering from SCD. This may facilitate personalized CV treatments.
Collapse
Affiliation(s)
- Eric J Niesor
- F.Hoffmann-La Roche Ltd, Basel, Switzerland. .,Pre-β1 Consulting, 13c Chemin de Bonmont, 1260, Nyon, Switzerland.
| | - Renée Benghozi
- F.Hoffmann-La Roche Ltd, Basel, Switzerland.,Cerenis Therapeutics Holding, Labège, France
| | | | | | | |
Collapse
|
356
|
Andersen MK, Sandholt CH. Recent Progress in the Understanding of Obesity: Contributions of Genome-Wide Association Studies. Curr Obes Rep 2015; 4:401-10. [PMID: 26374640 DOI: 10.1007/s13679-015-0173-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Since 2007, discovery of genetic variants associated with general obesity and fat distribution has advanced tremendously through genome-wide association studies (GWAS). Currently, the number of robustly associated loci is 190. Even though these loci explain <3 % of the variance, they have provided us a still emerging picture of genomic localization, frequency and effect size spectra, and hints of functional implications. The translation into biological knowledge has turned out to be an immense task. However, in silico enrichment analyses of genes involved in specific pathways or expressed in specific tissues have the power to suggest biological mechanisms underlying obesity. Inspired by this, we highlight genes in five loci potentially mechanistically linked to leptin-receptor trafficking and signaling in primary cilia. The clinical application of genetic knowledge as prediction, prevention, or treatment strategies is unfortunately still far from reality. Thus, despite major advances, further research is warranted to solve one of the greatest health problems in modern society.
Collapse
Affiliation(s)
- Mette Korre Andersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2.sal, 2100, Copenhagen, Denmark.
| | - Camilla Helene Sandholt
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2.sal, 2100, Copenhagen, Denmark.
| |
Collapse
|
357
|
Chesi A, Grant SFA. The Genetics of Pediatric Obesity. Trends Endocrinol Metab 2015; 26:711-721. [PMID: 26439977 PMCID: PMC4673034 DOI: 10.1016/j.tem.2015.08.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 01/24/2023]
Abstract
Obesity among children and adults has notably escalated over recent decades and represents a global major health problem. We now know that both genetic and environmental factors contribute to its complex etiology. Genome-wide association studies (GWAS) have revealed compelling genetic signals influencing obesity risk in adults. Recent reports for childhood obesity revealed that many adult loci also play a role in the pediatric setting. Childhood GWAS have uncovered novel loci below the detection range in adult studies, suggesting that obesity genes may be more easily uncovered in the pediatric setting. Shedding light on the genetic architecture of childhood obesity will facilitate the prevention and treatment of pediatric cases, and will have fundamental implications for diseases that present later in life.
Collapse
Affiliation(s)
- Alessandra Chesi
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Struan F A Grant
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
358
|
Gao C, Wang N, Guo X, Ziegler JT, Taylor KD, Xiang AH, Hai Y, Kridel SJ, Nadler JL, Kandeel F, Raffel LJ, Chen YDI, Norris JM, Rotter JI, Watanabe RM, Wagenknecht LE, Bowden DW, Speliotes EK, Goodarzi MO, Langefeld CD, Palmer ND. A Comprehensive Analysis of Common and Rare Variants to Identify Adiposity Loci in Hispanic Americans: The IRAS Family Study (IRASFS). PLoS One 2015; 10:e0134649. [PMID: 26599207 PMCID: PMC4658008 DOI: 10.1371/journal.pone.0134649] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/10/2015] [Indexed: 11/18/2022] Open
Abstract
Obesity is growing epidemic affecting 35% of adults in the United States. Previous genome-wide association studies (GWAS) have identified numerous loci associated with obesity. However, the majority of studies have been completed in Caucasians focusing on total body measures of adiposity. Here we report the results from genome-wide and exome chip association studies focusing on total body measures of adiposity including body mass index (BMI), percent body fat (PBF) and measures of fat deposition including waist circumference (WAIST), waist-hip ratio (WHR), subcutaneous adipose tissue (SAT), and visceral adipose tissue (VAT) in Hispanic Americans (nmax = 1263) from the Insulin Resistance Atherosclerosis Family Study (IRASFS). Five SNPs from two novel loci attained genome-wide significance (P<5.00x10-8) in IRASFS. A missense SNP in the isocitrate dehydrogenase 1 gene (IDH1) was associated with WAIST (rs34218846, MAF = 6.8%, PDOM = 1.62x10-8). This protein is postulated to play an important role in fat and cholesterol biosynthesis as demonstrated in cell and knock-out animal models. Four correlated intronic SNPs in the Zinc finger, GRF-type containing 1 gene (ZGRF1; SNP rs1471880, MAF = 48.1%, PDOM = 1.00x10-8) were strongly associated with WHR. The exact biological function of ZGRF1 and the connection with adiposity remains unclear. SNPs with p-values less than 5.00x10-6 from IRASFS were selected for replication. Meta-analysis was computed across seven independent Hispanic-American cohorts (nmax = 4156) and the strongest signal was rs1471880 (PDOM = 8.38x10-6) in ZGRF1 with WAIST. In conclusion, a genome-wide and exome chip association study was conducted that identified two novel loci (IDH1 and ZGRF1) associated with adiposity. While replication efforts were inconclusive, when taken together with the known biology, IDH1 and ZGRF1 warrant further evaluation.
Collapse
Affiliation(s)
- Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Nan Wang
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Julie T. Ziegler
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Kent D. Taylor
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Anny H. Xiang
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, United States of America
| | - Yang Hai
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Steven J. Kridel
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jerry L. Nadler
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Fouad Kandeel
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Leslie J. Raffel
- Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Yii-Der I. Chen
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Richard M. Watanabe
- Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Lynne E. Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Donald W. Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Elizabeth K. Speliotes
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mark O. Goodarzi
- Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Carl D. Langefeld
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Nicholette D. Palmer
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
359
|
Wells A, Kopp N, Xu X, O'Brien DR, Yang W, Nehorai A, Adair-Kirk TL, Kopan R, Dougherty JD. The anatomical distribution of genetic associations. Nucleic Acids Res 2015; 43:10804-20. [PMID: 26586807 PMCID: PMC4678833 DOI: 10.1093/nar/gkv1262] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 11/04/2015] [Indexed: 01/13/2023] Open
Abstract
Deeper understanding of the anatomical intermediaries for disease and other complex genetic traits is essential to understanding mechanisms and developing new interventions. Existing ontology tools provide functional, curated annotations for many genes and can be used to develop mechanistic hypotheses; yet information about the spatial expression of genes may be equally useful in interpreting results and forming novel hypotheses for a trait. Therefore, we developed an approach for statistically testing the relationship between gene expression across the body and sets of candidate genes from across the genome. We validated this tool and tested its utility on three applications. First, we show that the expression of genes in associated loci from GWA studies implicates specific tissues for 57 out of 98 traits. Second, we tested the ability of the tool to identify novel relationships between gene expression and phenotypes. Specifically, we experimentally confirmed an underappreciated prediction highlighted by our tool: that white blood cell count--a quantitative trait of the immune system--is genetically modulated by genes expressed in the skin. Finally, using gene lists derived from exome sequencing data, we show that human genes under selective constraint are disproportionately expressed in nervous system tissues.
Collapse
Affiliation(s)
- Alan Wells
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nathan Kopp
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaoxiao Xu
- The Preston M. Green Department of Electrical and Systems Engineering, Washington University, St. Louis, MO 63130, USA
| | - David R O'Brien
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Arye Nehorai
- The Preston M. Green Department of Electrical and Systems Engineering, Washington University, St. Louis, MO 63130, USA
| | - Tracy L Adair-Kirk
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Raphael Kopan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - J D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
360
|
Cooper GF, Bahar I, Becich MJ, Benos PV, Berg J, Espino JU, Glymour C, Jacobson RC, Kienholz M, Lee AV, Lu X, Scheines R. The center for causal discovery of biomedical knowledge from big data. J Am Med Inform Assoc 2015; 22:1132-6. [PMID: 26138794 PMCID: PMC5009908 DOI: 10.1093/jamia/ocv059] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 04/27/2015] [Accepted: 05/02/2015] [Indexed: 01/12/2023] Open
Abstract
The Big Data to Knowledge (BD2K) Center for Causal Discovery is developing and disseminating an integrated set of open source tools that support causal modeling and discovery of biomedical knowledge from large and complex biomedical datasets. The Center integrates teams of biomedical and data scientists focused on the refinement of existing and the development of new constraint-based and Bayesian algorithms based on causal Bayesian networks, the optimization of software for efficient operation in a supercomputing environment, and the testing of algorithms and software developed using real data from 3 representative driving biomedical projects: cancer driver mutations, lung disease, and the functional connectome of the human brain. Associated training activities provide both biomedical and data scientists with the knowledge and skills needed to apply and extend these tools. Collaborative activities with the BD2K Consortium further advance causal discovery tools and integrate tools and resources developed by other centers.
Collapse
Affiliation(s)
- Gregory F Cooper
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael J Becich
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeremy Berg
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA Institute for Personalized Medicine, University of Pittsburgh and University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Jeremy U Espino
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Clark Glymour
- Department of Philosophy, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Michelle Kienholz
- Institute for Personalized Medicine, University of Pittsburgh and University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Adrian V Lee
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Richard Scheines
- Dietrich College of Humanities and Social Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
361
|
Kim J, Bai Y, Pan W. An Adaptive Association Test for Multiple Phenotypes with GWAS Summary Statistics. Genet Epidemiol 2015; 39:651-63. [PMID: 26493956 DOI: 10.1002/gepi.21931] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023]
Abstract
We study the problem of testing for single marker-multiple phenotype associations based on genome-wide association study (GWAS) summary statistics without access to individual-level genotype and phenotype data. For most published GWASs, because obtaining summary data is substantially easier than accessing individual-level phenotype and genotype data, while often multiple correlated traits have been collected, the problem studied here has become increasingly important. We propose a powerful adaptive test and compare its performance with some existing tests. We illustrate its applications to analyses of a meta-analyzed GWAS dataset with three blood lipid traits and another with sex-stratified anthropometric traits, and further demonstrate its potential power gain over some existing methods through realistic simulation studies. We start from the situation with only one set of (possibly meta-analyzed) genome-wide summary statistics, then extend the method to meta-analysis of multiple sets of genome-wide summary statistics, each from one GWAS. We expect the proposed test to be useful in practice as more powerful than or complementary to existing methods.
Collapse
Affiliation(s)
- Junghi Kim
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Yun Bai
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
362
|
Kim K, Ahn N, Park J, Koh J, Jung S, Kim S, Moon S. Association of angiotensin-converting enzyme I/D and α-actinin-3 R577X genotypes with metabolic syndrome risk factors in Korean children. Obes Res Clin Pract 2015; 10 Suppl 1:S125-S132. [PMID: 26483160 DOI: 10.1016/j.orcp.2015.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/29/2015] [Accepted: 09/18/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE This study analysed the risk factors associated with metabolic syndrome through the interaction between ACTN3 and ACE gene polymorphism in Korean children. METHODS The subjects of the study consisted of elementary school students (n=788, age 10.10±0.07 yr). The anthropometric parameters, blood lipid profiles, and metabolic markers were compared among groups of the ACE I/D or the ACTN3 R577X polymorphisms. RESULTS The subjects with the DD genotype showed significantly higher systolic blood pressure than the subjects with the II and ID genotype of the ACE gene polymorphism. XX genotype had significantly lower waist-hip ratio than those with RR genotype of the ACTN3 gene polymorphism. Also, the subjects with XX genotype exhibited significantly higher blood HDL cholesterol level than those with RR or RX genotype. The interaction of ACTN3 and ACE gene polymorphism in subjects having both ACE DD and ACTN3 RR genotypes demonstrated a significantly higher metabolic syndrome score than any other groups. CONCLUSION The children having both ACTN3 RR or RX genotype and ACE DD genotype showed high systolic blood pressure and low blood HDL cholesterol level, which may be considered a high-risk in metabolic syndrome.
Collapse
Affiliation(s)
- Kijin Kim
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu, Republic of Korea.
| | - Nayoung Ahn
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu, Republic of Korea
| | - Jusik Park
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu, Republic of Korea
| | - Jinho Koh
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu, Republic of Korea
| | - Suryun Jung
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu, Republic of Korea
| | - Sanghyun Kim
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu, Republic of Korea
| | - Sangbok Moon
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
363
|
Karaderi T, Drong AW, Lindgren CM. Insights into the Genetic Susceptibility to Type 2 Diabetes from Genome-Wide Association Studies of Obesity-Related Traits. Curr Diab Rep 2015; 15:83. [PMID: 26363598 PMCID: PMC4568008 DOI: 10.1007/s11892-015-0648-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Obesity and type 2 diabetes (T2D) are common and complex metabolic diseases, which are caused by an interchange between environmental and genetic factors. Recently, a number of large-scale genome-wide association studies (GWAS) have improved our knowledge of the genetic architecture and biological mechanisms of these diseases. Currently, more than ~250 genetic loci have been found for monogenic, syndromic, or common forms of T2D and/or obesity-related traits. In this review, we discuss the implications of these GWAS for obesity and T2D, and investigate the overlap of loci for obesity-related traits and T2D, highlighting potential mechanisms that affect T2D susceptibility.
Collapse
Affiliation(s)
- Tugce Karaderi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, OX3 7BN, Oxford, UK.
| | - Alexander W Drong
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, OX3 7BN, Oxford, UK.
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, OX3 7BN, Oxford, UK.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Big Data Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
364
|
Asimit JL, Panoutsopoulou K, Wheeler E, Berndt SI, Cordell HJ, Morris AP, Zeggini E, Barroso I. A Bayesian Approach to the Overlap Analysis of Epidemiologically Linked Traits. Genet Epidemiol 2015; 39:624-34. [PMID: 26411566 PMCID: PMC4832282 DOI: 10.1002/gepi.21919] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 06/04/2015] [Accepted: 07/20/2015] [Indexed: 01/08/2023]
Abstract
Diseases often cooccur in individuals more often than expected by chance, and may be explained by shared underlying genetic etiology. A common approach to genetic overlap analyses is to use summary genome‐wide association study data to identify single‐nucleotide polymorphisms (SNPs) that are associated with multiple traits at a selected P‐value threshold. However, P‐values do not account for differences in power, whereas Bayes’ factors (BFs) do, and may be approximated using summary statistics. We use simulation studies to compare the power of frequentist and Bayesian approaches with overlap analyses, and to decide on appropriate thresholds for comparison between the two methods. It is empirically illustrated that BFs have the advantage over P‐values of a decreasing type I error rate as study size increases for single‐disease associations. Consequently, the overlap analysis of traits from different‐sized studies encounters issues in fair P‐value threshold selection, whereas BFs are adjusted automatically. Extensive simulations show that Bayesian overlap analyses tend to have higher power than those that assess association strength with P‐values, particularly in low‐power scenarios. Calibration tables between BFs and P‐values are provided for a range of sample sizes, as well as an approximation approach for sample sizes that are not in the calibration table. Although P‐values are sometimes thought more intuitive, these tables assist in removing the opaqueness of Bayesian thresholds and may also be used in the selection of a BF threshold to meet a certain type I error rate. An application of our methods is used to identify variants associated with both obesity and osteoarthritis.
Collapse
Affiliation(s)
| | | | - Eleanor Wheeler
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Heather J Cordell
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,Department of Biostatistics, University of Liverpool, Liverpool, United Kingdom
| | | | - Inês Barroso
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| |
Collapse
|
365
|
Garton FC, North KN, Koch LG, Britton SL, Nogales-Gadea G, Lucia A. Rodent models for resolving extremes of exercise and health. Physiol Genomics 2015; 48:82-92. [PMID: 26395598 DOI: 10.1152/physiolgenomics.00077.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The extremes of exercise capacity and health are considered a complex interplay between genes and the environment. In general, the study of animal models has proven critical for deep mechanistic exploration that provides guidance for focused and hypothesis-driven discovery in humans. Hypotheses underlying molecular mechanisms of disease and gene/tissue function can be tested in rodents to generate sufficient evidence to resolve and progress our understanding of human biology. Here we provide examples of three alternative uses of rodent models that have been applied successfully to advance knowledge that bridges our understanding of the connection between exercise capacity and health status. First we review the strong association between exercise capacity and all-cause morbidity and mortality in humans through artificial selection on low and high exercise performance in the rat and the consequent generation of the "energy transfer hypothesis." Second we review specific transgenic and knockout mouse models that replicate the human disease condition and performance. This includes human glycogen storage diseases (McArdle and Pompe) and α-actinin-3 deficiency. Together these rodent models provide an overview of the advancements of molecular knowledge required for clinical translation. Continued study of these models in conjunction with human association studies will be critical to resolving the complex gene-environment interplay linking exercise capacity, health, and disease.
Collapse
Affiliation(s)
- Fleur C Garton
- Murdoch Childrens Research Institute, Melbourne, Victoria, Australia; Royal Children's Hospital, Department of Paediatrics, Melbourne, Victoria, Australia;
| | - Kathryn N North
- Murdoch Childrens Research Institute, Melbourne, Victoria, Australia; Royal Children's Hospital, Department of Paediatrics, Melbourne, Victoria, Australia
| | - Lauren G Koch
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Gisela Nogales-Gadea
- Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol i Campus Can Ruti, Universitat Autònoma de Barcelona, Badalona, Spain; and
| | - Alejandro Lucia
- Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol i Campus Can Ruti, Universitat Autònoma de Barcelona, Badalona, Spain; and Instituto de Investigación Hospital 12 de Octubre (i+12) and Universidad Europea, Madrid, Spain
| |
Collapse
|
366
|
Perspectives on pharmacogenomics of antiretroviral medications and HIV-associated comorbidities. Curr Opin HIV AIDS 2015; 10:116-22. [PMID: 25565175 DOI: 10.1097/coh.0000000000000134] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW To summarize current knowledge and provide perspective on relationships between human genetic variants, antiretroviral medications, and aging-related complications of HIV-1 infection. RECENT FINDINGS Human genetic variants have been convincingly associated with interindividual variability in antiretroviral toxicities, drug disposition, and aging-associated complications in HIV-1 infection. Screening for HLA-B5701 to avoid abacavir hypersensitivity reactions has become a routine part of clinical care, and has markedly improved drug safety. There are well established pharmacogenetic associations with other agents (efavirenz, nevirapine, atazanavir, dolutegravir, and others), but this knowledge has yet to have substantial impact on HIV-1 clinical care. As metabolic complications including diabetes mellitus, dyslipidemia, osteoporosis, and cardiovascular disease are becoming an increasing concern among individuals who are aging with well controlled HIV-1 infection, human genetic variants that predispose to these complications also become more relevant in this population. SUMMARY Pharmacogenetic knowledge has already had considerable impact on antiretroviral prescribing. With continued advances in the field of human genomics, the impact of pharmacogenomics on HIV-1 clinical care and research is likely to continue to grow in importance and scope.
Collapse
|
367
|
Soileau L, Bautista D, Johnson C, Gao C, Zhang K, Li X, Heymsfield SB, Thomas D, Zheng J. Automated anthropometric phenotyping with novel Kinect-based three-dimensional imaging method: comparison with a reference laser imaging system. Eur J Clin Nutr 2015; 70:475-81. [PMID: 26373966 DOI: 10.1038/ejcn.2015.132] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/25/2015] [Accepted: 07/14/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND/OBJECTIVES Anthropometry for measuring body composition, shape, surface area and volume is important for human clinical research and practice. Although training and technical skills are required for traditional tape and caliper anthropometry, a new opportunity exists for automated measurement using newly developed relatively low-cost three-dimensional (3D) imaging devices. The aim of this study was to compare results provided by a Kinect-based device to a traditional laser 3D reference system. SUBJECTS/METHODS Measurements made by the evaluated device, a hybrid of commercially purchased hardware (KX-16; TC(2), Cary, NC, USA) with our additional added software, were compared with those derived by a high-resolution laser scanner (Vitus Smart XXL; Human Solutions North America, Cary, NC, USA). Both imaging systems were compared with additional linear (stadiometer-derived height) and volumetric (total volume, air-displacement plethysmography) measurements. Subjects (n=101) were healthy children (age ≥5 years) and adults varying in body mass index. RESULTS Representative linear (4), circumferential (6), volumetric (3) and surface area (1) measurements made by the Kinect-based device showed a consistent pattern relative to the laser system: high correlations (R(2)s= 0.70-0.99, all P<0.001); 1-3% differences for large linear (for example, height, X±s.d., -1.4±0.5%), circumferential (for example, waist circumference, -2.1±1.8%), volume (for example, total body, -0.8±2.2%) and surface area (whole-body, -1.7±2.0%) estimates. By contrast, mean measurement differences were substantially larger for small structures (for example, forearm volume, 31.3±31.4%). CONCLUSIONS Low-cost 3D Kinect-based imaging systems have the potential for providing automated accurate anthropometric and related body measurements for relatively large components; further hardware and software developments may be able to improve system small-component resolution.
Collapse
Affiliation(s)
- L Soileau
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, USA
| | - D Bautista
- Tulane University School of Medicine, New Orleans, LA, USA
| | - C Johnson
- Body Composition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - C Gao
- Body Composition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - K Zhang
- Division of Electrical and Computer Engineering, School of Electrical Engineering and Computer Science, Louisiana State University, Baton Rouge, LA, USA
| | - X Li
- Division of Electrical and Computer Engineering, School of Electrical Engineering and Computer Science, Louisiana State University, Baton Rouge, LA, USA
| | - S B Heymsfield
- Body Composition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - D Thomas
- Department of Mathematics, Center for Quantitative Obesity Research, Montclair State University, Montclair, NJ, USA
| | - J Zheng
- Body Composition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
368
|
Pant SD, Karlskov-Mortensen P, Jacobsen MJ, Cirera S, Kogelman LJA, Bruun CS, Mark T, Jørgensen CB, Grarup N, Appel EVR, Galjatovic EAA, Hansen T, Pedersen O, Guerin M, Huby T, Lesnik P, Meuwissen THE, Kadarmideen HN, Fredholm M. Comparative Analyses of QTLs Influencing Obesity and Metabolic Phenotypes in Pigs and Humans. PLoS One 2015; 10:e0137356. [PMID: 26348622 PMCID: PMC4562524 DOI: 10.1371/journal.pone.0137356] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/14/2015] [Indexed: 12/31/2022] Open
Abstract
The pig is a well-known animal model used to investigate genetic and mechanistic aspects of human disease biology. They are particularly useful in the context of obesity and metabolic diseases because other widely used models (e.g. mice) do not completely recapitulate key pathophysiological features associated with these diseases in humans. Therefore, we established a F2 pig resource population (n = 564) designed to elucidate the genetics underlying obesity and metabolic phenotypes. Segregation of obesity traits was ensured by using breeds highly divergent with respect to obesity traits in the parental generation. Several obesity and metabolic phenotypes were recorded (n = 35) from birth to slaughter (242 ± 48 days), including body composition determined at about two months of age (63 ± 10 days) via dual-energy x-ray absorptiometry (DXA) scanning. All pigs were genotyped using Illumina Porcine 60k SNP Beadchip and a combined linkage disequilibrium-linkage analysis was used to identify genome-wide significant associations for collected phenotypes. We identified 229 QTLs which associated with adiposity- and metabolic phenotypes at genome-wide significant levels. Subsequently comparative analyses were performed to identify the extent of overlap between previously identified QTLs in both humans and pigs. The combined analysis of a large number of obesity phenotypes has provided insight in the genetic architecture of the molecular mechanisms underlying these traits indicating that QTLs underlying similar phenotypes are clustered in the genome. Our analyses have further confirmed that genetic heterogeneity is an inherent characteristic of obesity traits most likely caused by segregation or fixation of different variants of the individual components belonging to cellular pathways in different populations. Several important genes previously associated to obesity in human studies, along with novel genes were identified. Altogether, this study provides novel insight that may further the current understanding of the molecular mechanisms underlying human obesity.
Collapse
Affiliation(s)
- Sameer D. Pant
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Karlskov-Mortensen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette J. Jacobsen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanna Cirera
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisette J. A. Kogelman
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla S. Bruun
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Mark
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus B. Jørgensen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emil V. R. Appel
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ehm A. A. Galjatovic
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maryse Guerin
- INSERM UMR_S 1166, Integrative Biology of Atherosclerosis Team, F-75013, Paris, France
- Sorbonne Universités UPMC Univ Paris 06 UMR_S 1166, Integrative Biology of Atherosclerosis Team, F-75013, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Thierry Huby
- INSERM UMR_S 1166, Integrative Biology of Atherosclerosis Team, F-75013, Paris, France
- Sorbonne Universités UPMC Univ Paris 06 UMR_S 1166, Integrative Biology of Atherosclerosis Team, F-75013, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Philipppe Lesnik
- INSERM UMR_S 1166, Integrative Biology of Atherosclerosis Team, F-75013, Paris, France
- Sorbonne Universités UPMC Univ Paris 06 UMR_S 1166, Integrative Biology of Atherosclerosis Team, F-75013, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Theo H. E. Meuwissen
- Institute of Animal and Agricultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Haja N. Kadarmideen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (MF); (HNK)
| | - Merete Fredholm
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (MF); (HNK)
| |
Collapse
|
369
|
Ashraf T, Collinson MN, Fairhurst J, Wang R, Wilson LC, Foulds N. Two further patients with the 1q24 deletion syndrome expand the phenotype: A possible role for the miR199-214 cluster in the skeletal features of the condition. Am J Med Genet A 2015; 167A:3153-60. [PMID: 26333682 DOI: 10.1002/ajmg.a.37336] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 08/02/2015] [Indexed: 01/02/2023]
Abstract
Submicroscopic deletions within chromosome 1q24q25 are associated with a syndromic phenotype of short stature, brachydactyly, learning difficulties, and facial dysmorphism. The critical region for the deletion phenotype has previously been narrowed to a 1.9 Mb segment containing 13 genes. We describe two further patients with 1q24 microdeletions and the skeletal phenotype, the first of whom has normal intellect, whereas the second has only mild learning impairment. The deletion in the first patient is very small and further narrows the critical interval for the striking skeletal aspects of this condition to a region containing only Dynamin 3 (DNM3) and two microRNAs that are harbored within intron 14 of this gene: miR199 and miR214. Mouse studies raise the possibility that these microRNAs may be implicated in the short stature and skeletal abnormalities of this microdeletion condition. The deletion in the second patient spans the previously reported critical region and indicates that the cognitive impairment may not always be as severe as previous reports suggest.
Collapse
Affiliation(s)
- Tazeen Ashraf
- Guy's Clinical Genetics Service, Guy's Hospital, London, United Kingdom
| | - Morag N Collinson
- Wessex Regional Genetics Laboratory, Salisbury NHS Foundation Trust, Salisbury, Wiltshire, United Kingdom
| | - Joanna Fairhurst
- Radiology Department, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, United Kingdom
| | - Rubin Wang
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Louise C Wilson
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Nicola Foulds
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, United Kingdom
| |
Collapse
|
370
|
Wu B, Pankow JS, Guan W. Sequence Kernel Association Analysis of Rare Variant Set Based on the Marginal Regression Model for Binary Traits. Genet Epidemiol 2015; 39:399-405. [PMID: 26282996 PMCID: PMC4544778 DOI: 10.1002/gepi.21913] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 01/28/2023]
Abstract
Recent sequencing efforts have focused on exploring the influence of rare variants on the complex diseases. Gene level based tests by aggregating information across rare variants within a gene have become attractive to enrich the rare variant association signal. Among them, the sequence kernel association test (SKAT) has proved to be a very powerful method for jointly testing multiple rare variants within a gene. In this article, we explore an alternative SKAT. We propose to use the univariate likelihood ratio statistics from the marginal model for individual variants as input into the kernel association test. We show how to compute its significance P-value efficiently based on the asymptotic chi-square mixture distribution. We demonstrate through extensive numerical studies that the proposed method has competitive performance. Its usefulness is further illustrated with application to associations between rare exonic variants and type 2 diabetes (T2D) in the Atherosclerosis Risk in Communities (ARIC) study. We identified an exome-wide significant rare variant set in the gene ZZZ3 worthy of further investigations.
Collapse
Affiliation(s)
- Baolin Wu
- Division of Biostatistics, School of Public Health, University of
Minnesota
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public
Health, University of Minnesota
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of
Minnesota
| |
Collapse
|
371
|
Pathway analysis of body mass index genome-wide association study highlights risk pathways in cardiovascular disease. Sci Rep 2015; 5:13025. [PMID: 26264282 PMCID: PMC4533004 DOI: 10.1038/srep13025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/15/2015] [Indexed: 01/02/2023] Open
Abstract
Cardiovascular disease (CVD) is a class of diseases that involve the heart or blood vessels. It is reported that body mass index (BMI) is risk factor for CVD. Genome-wide association studies (GWAS) have recently provided rapid insights into genetics of CVD and its risk factors. However, the specific mechanisms how BMI influences CVD risk are largely unknown. We think that BMI may influences CVD risk by shared genetic pathways. In order to confirm this view, we conducted a pathway analysis of BMI GWAS, which examined approximately 329,091 single nucleotide polymorphisms from 4763 samples. We identified 31 significant KEGG pathways. There is literature evidence supporting the involvement of GnRH signaling, vascular smooth muscle contraction, dilated cardiomyopathy, Gap junction, Wnt signaling, Calcium signaling and Chemokine signaling in CVD. Collectively, our study supports the potential role of the CVD risk pathways in BMI. BMI may influence CVD risk by the shared genetic pathways. We believe that our results may advance our understanding of BMI mechanisms in CVD.
Collapse
|
372
|
Tallapragada DSP, Bhaskar S, Chandak GR. New insights from monogenic diabetes for "common" type 2 diabetes. Front Genet 2015; 6:251. [PMID: 26300908 PMCID: PMC4528293 DOI: 10.3389/fgene.2015.00251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/13/2015] [Indexed: 01/17/2023] Open
Abstract
Boundaries between monogenic and complex genetic diseases are becoming increasingly blurred, as a result of better understanding of phenotypes and their genetic determinants. This had a large impact on the way complex disease genetics is now being investigated. Starting with conventional approaches like familial linkage, positional cloning and candidate genes strategies, the scope of complex disease genetics has grown exponentially with scientific and technological advances in recent times. Despite identification of multiple loci harboring common and rare variants associated with complex diseases, interpreting and evaluating their functional role has proven to be difficult. Information from monogenic diseases, especially related to the intermediate traits associated with complex diseases comes handy. The significant overlap between traits and phenotypes of monogenic diseases with related complex diseases provides a platform to understand the disease biology better. In this review, we would discuss about one such complex disease, type 2 diabetes, which shares marked similarity of intermediate traits with different forms of monogenic diabetes.
Collapse
Affiliation(s)
| | | | - Giriraj R. Chandak
- Genomic Research on Complex Diseases Laboratory, Council of Scientific and Industrial Research-Centre for Cellular and Molecular BiologyHyderabad, India
| |
Collapse
|
373
|
Abstract
There is ongoing controversy as to whether obesity confers risk for CAD independently of associated risk factors including diabetes mellitus. We have carried out a Mendelian randomization study using a genetic risk score (GRS) for body mass index (BMI) based on 35 risk alleles to investigate this question in a population of 5831 early onset CAD cases without diabetes mellitus and 3832 elderly healthy control subjects, all of strictly European ancestry, with adjustment for traditional risk factors (TRFs). We then estimated the genetic correlation between these BMI and CAD (rg) by relating the pairwise genetic similarity matrix to a phenotypic covariance matrix between these two traits. GRSBMI significantly (P=2.12 × 10−12) associated with CAD status in a multivariate model adjusted for TRFs, with a per allele odds ratio (OR) of 1.06 (95% CI 1.042–1.076). The addition of GRSBMI to TRFs explained 0.75% of CAD variance and yielded a continuous net recombination index of 16.54% (95% CI=11.82–21.26%, P<0.0001). To test whether GRSBMI explained CAD status when adjusted for measured BMI, separate models were constructed in which the score and BMI were either included as covariates or not. The addition of BMI explained ~1.9% of CAD variance and GRSBMI plus BMI explained 2.65% of CAD variance. Finally, using bivariate restricted maximum likelihood analysis, we provide strong evidence of genome-wide pleiotropy between obesity and CAD. This analysis supports the hypothesis that obesity is a causal risk factor for CAD.
Collapse
|
374
|
Crawford DC, Goodloe R, Farber-Eger E, Boston J, Pendergrass SA, Haines JL, Ritchie MD, Bush WS. Leveraging Epidemiologic and Clinical Collections for Genomic Studies of Complex Traits. Hum Hered 2015. [PMID: 26201699 DOI: 10.1159/000381805] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND/AIMS Present-day limited resources demand DNA and phenotyping alternatives to the traditional prospective population-based epidemiologic collections. METHODS To accelerate genomic discovery with an emphasis on diverse populations, we--as part of the Epidemiologic Architecture for Genes Linked to Environment (EAGLE) study--accessed all non-European American samples (n = 15,863) available in BioVU, the Vanderbilt University biorepository linked to de-identified electronic medical records, for genomic studies as part of the larger Population Architecture using Genomics and Epidemiology (PAGE) I study. Given previous studies have cautioned against the secondary use of clinically collected data compared with epidemiologically collected data, we present here a characterization of EAGLE BioVU, including the billing and diagnostic (ICD-9) code distributions for adult and pediatric patients as well as comparisons made for select health metrics (body mass index, glucose, HbA1c, HDL-C, LDL-C, and triglycerides) with the population-based National Health and Nutrition Examination Surveys (NHANES) linked to DNA samples (NHANES III, n = 7,159; NHANES 1999-2002, n = 7,839). RESULTS Overall, the distributions of billing and diagnostic codes suggest this clinical sample is a mixture of healthy and sick patients like that expected for a contemporary American population. CONCLUSION Little bias is observed among health metrics, suggesting this clinical collection is suitable for genomic studies along with traditional epidemiologic cohorts.
Collapse
Affiliation(s)
- Dana C Crawford
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | |
Collapse
|
375
|
Dos Santos FK, Nevill A, Gomes TNQF, Chaves R, Daca T, Madeira A, Katzmarzyk PT, Prista A, Maia JAR. Differences in motor performance between children and adolescents in Mozambique and Portugal: impact of allometric scaling. Ann Hum Biol 2015. [PMID: 26207594 DOI: 10.3109/03014460.2015.1024738] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Children from developed and developing countries have different anthropometric characteristics which may affect their motor performance (MP). AIM To use the allometric approach to model the relationship between body size and MP in youth from two countries differing in socio-economic status-Portugal and Mozambique. SUBJECTS AND METHODS A total of 2946 subjects, 1280 Mozambicans (688 girls) and 1666 Portuguese (826 girls), aged 10-15 years were sampled. Height and weight were measured and the reciprocal ponderal index (RPI) was computed. MP included handgrip strength, 1-mile run/walk, curl-ups and standing long jump tests. A multiplicative allometric model was adopted to adjust for body size differences across countries. RESULTS Differences in MP between Mozambican and Portuguese children exist, invariably favouring the latter. The allometric models used to adjust MP for differences in body size identified the optimal body shape to be either the RPI or even more linear, i.e. approximately (height/mass(0.25)). Having adjusted the MP variables for differences in body size, the differences between Mozambican and Portuguese children were invariably reduced and, in the case of grip strength, reversed. CONCLUSION These results reinforce the notion that significant differences exist in MP across countries, even after adjusting for differences in body size.
Collapse
Affiliation(s)
- Fernanda Karina Dos Santos
- a CIFI2D, Kinanthropometry Lab, Faculty of Sport, University of Porto , Porto , Portugal .,b CAPES Foundation, Ministry of Education of Brazil , Brasília - DF , Brazil
| | - Allan Nevill
- c School of Sport, Performing Arts and Leisure, University of Wolverhampton , Walsall , UK
| | | | - Raquel Chaves
- d Federal University of Technology - Paraná (UFTFPR), Campus Curitiba , Curitiba , Brazil
| | - Timóteo Daca
- e Faculty of Physical Education and Sports , Pedagogical University , Maputo , Mozambique , and
| | - Aspacia Madeira
- e Faculty of Physical Education and Sports , Pedagogical University , Maputo , Mozambique , and
| | - Peter T Katzmarzyk
- f Pennington Biomedical Research Center, Louisiana State University , Baton Rouge , LA , USA
| | - António Prista
- e Faculty of Physical Education and Sports , Pedagogical University , Maputo , Mozambique , and
| | - José A R Maia
- a CIFI2D, Kinanthropometry Lab, Faculty of Sport, University of Porto , Porto , Portugal
| |
Collapse
|
376
|
Yao P, Lin P, Gokoolparsadh A, Assareh A, Thang MWC, Voineagu I. Coexpression networks identify brain region-specific enhancer RNAs in the human brain. Nat Neurosci 2015; 18:1168-74. [PMID: 26167905 DOI: 10.1038/nn.4063] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/18/2015] [Indexed: 12/17/2022]
Abstract
Despite major progress in identifying enhancer regions on a genome-wide scale, the majority of available data are limited to model organisms and human transformed cell lines. We have identified a robust set of enhancer RNAs (eRNAs) expressed in the human brain and constructed networks assessing eRNA-gene coexpression interactions across human fetal brain and multiple adult brain regions. Our data identify brain region-specific eRNAs and show that enhancer regions expressing eRNAs are enriched for genetic variants associated with autism spectrum disorders.
Collapse
Affiliation(s)
- Pu Yao
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Peijie Lin
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Akira Gokoolparsadh
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Amelia Assareh
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Mike W C Thang
- QFAB Bioinformatics, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Irina Voineagu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
377
|
Giri A, Edwards TL, Motley SS, Byerly SH, Fowke JH. Genetic Determinants of Metabolism and Benign Prostate Enlargement: Associations with Prostate Volume. PLoS One 2015; 10:e0132028. [PMID: 26158673 PMCID: PMC4497718 DOI: 10.1371/journal.pone.0132028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/09/2015] [Indexed: 11/18/2022] Open
Abstract
Prostate enlargement leading to clinical benign prostatic hyperplasia (BPH) is associated with metabolic dysregulation and obesity. The genetic basis of this association is unclear. Our objective was to evaluate whether single nucleotide polymorphisms (SNPs) previously associated with metabolic disorders are also associated with prostate volume (PV). Participants included 876 men referred for prostate biopsy and found to be prostate cancer free. PV was measured by transrectal ultrasound. Samples were genotyped using the Illumina Cardio-MetaboChip platform. Multivariable adjusted linear regression models were used to evaluate SNPs (additive coding) in relation to natural-log transformed (log) PV. We compared SNP-PV results from biopsy-negative men to 442 men with low-grade prostate cancer with similar levels of obesity and PV. Beta-coefficients from the discovery and replication samples were then aggregated with fixed effects inverse variance weighted meta-analysis. SNP rs11736129 (near the pseudo-gene LOC100131429) was significantly associated with log-PV (beta: 0.16, p-value 1.16x10-8) after adjusting for multiple testing. Other noteworthy SNPs that were nominally associated (p-value < 1x10-4) with log-PV included rs9583484 (intronic SNP in COL4A2), rs10146527 (intronic SNP in NRXN3), rs9909466 (SNP near RPL32P31), and rs2241606 (synonymous SNP in SLC12A7). We found several SNPs in metabolic loci associated with PV. Further studies are needed to confirm our results and elucidate the mechanism between these genetic loci, PV, and clinical BPH.
Collapse
Affiliation(s)
- Ayush Giri
- Institute for Medicine and Public Health, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Todd L. Edwards
- Institute for Medicine and Public Health, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, United States of America
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Saundra S. Motley
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Susan H. Byerly
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Jay H. Fowke
- Institute for Medicine and Public Health, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Surgical Urology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
378
|
Pharmacogenomic variants have larger effect sizes than genetic variants associated with other dichotomous complex traits. THE PHARMACOGENOMICS JOURNAL 2015; 16:388-92. [PMID: 26149738 PMCID: PMC4704992 DOI: 10.1038/tpj.2015.47] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/17/2015] [Accepted: 05/21/2015] [Indexed: 01/15/2023]
Abstract
It has been suggested that pharmacogenomic phenotypes are influenced by genetic variants with larger effect sizes than other phenotypes, such as complex disease risk. This is presumed to reflect the fact that relevant environmental factors (drug exposure) are appropriately measured and taken into account. To test this hypothesis, we performed a systematic comparison of effect sizes between pharmacogenomic and non-pharmacogenomic phenotypes across all genome-wide association studies (GWAS) reported in the NHGRI GWAS catalog. We found significantly larger effect sizes for studies focused on pharmacogenomic phenotypes, as compared to complex disease risk, morphological phenotypes, and endophenotypes. We found no significant differences in effect sizes between pharmacogenomic studies focused on adverse events versus those focused on drug efficacy. Furthermore, we found that this pattern persists among sample size-matched studies, suggesting that this pattern does not reflect over-estimation of effect sizes due to smaller sample sizes in pharmacogenomic studies.
Collapse
|
379
|
Viljakainen H, Andersson-Assarsson JC, Armenio M, Pekkinen M, Pettersson M, Valta H, Lipsanen-Nyman M, Mäkitie O, Lindstrand A. Low Copy Number of the AMY1 Locus Is Associated with Early-Onset Female Obesity in Finland. PLoS One 2015; 10:e0131883. [PMID: 26132294 PMCID: PMC4489572 DOI: 10.1371/journal.pone.0131883] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/08/2015] [Indexed: 12/25/2022] Open
Abstract
Background The salivary α-amylase locus (AMY1) is located in a highly polymorphic multi allelic copy number variable chromosomal region. A recent report identified an association between AMY1 copy numbers and BMI in common obesity. The present study investigated the relationship between AMY1 copy number, BMI and serum amylase in childhood-onset obesity. Patients Sixty-one subjects with a history of childhood-onset obesity (mean age 19.1 years, 54% males) and 71 matched controls (19.8 yrs, 45% males) were included. All anthropometric measures were greater in the obese; their mean BMI was 40 kg/m2 (range 25-62 kg/m2) compared with 23 kg/m2 in the controls (15-32 kg/m2). Results Mean AMY1 copy numbers did not differ between the obese and control subjects, but gender differences were observed; obese men showed the highest and obese women the lowest number of AMY1 copies (p=0.045). Further, only in affected females, AMY1 copy number correlated significantly with whole body fat percent (r=-0.512, p=0.013) and BMI (r=-0.416, p=0.025). Finally, a clear linear association between AMY1 copy number and serum salivary amylase was observed in all subgroups but again differences existed between obese males and females. Conclusions In conclusion, our findings suggest that AMY1 copy number differences play a role in childhood-onset obesity but the effect differs between males and females. Further studies in larger cohorts are needed to confirm these observations.
Collapse
Affiliation(s)
- Heli Viljakainen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johanna C Andersson-Assarsson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Center for Cardiovascular and Metabolic Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Miriam Armenio
- Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Maria Pettersson
- Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Helena Valta
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Marita Lipsanen-Nyman
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Outi Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
380
|
Genetics of second-generation antipsychotic and mood stabilizer-induced weight gain in bipolar disorder. Pharmacogenet Genomics 2015; 25:354-62. [DOI: 10.1097/fpc.0000000000000144] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
381
|
Lappalainen I, Almeida-King J, Kumanduri V, Senf A, Spalding JD, Ur-Rehman S, Saunders G, Kandasamy J, Caccamo M, Leinonen R, Vaughan B, Laurent T, Rowland F, Marin-Garcia P, Barker J, Jokinen P, Torres AC, de Argila JR, Llobet OM, Medina I, Puy MS, Alberich M, de la Torre S, Navarro A, Paschall J, Flicek P. The European Genome-phenome Archive of human data consented for biomedical research. Nat Genet 2015; 47:692-5. [PMID: 26111507 PMCID: PMC5426533 DOI: 10.1038/ng.3312] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The European Genome-phenome Archive (EGA) is a permanent archive that promotes distribution and sharing of genetic and phenotype data consented for specific approved uses, but not fully open public distribution. The EGA follows strict protocols for information management, data storage, security and dissemination. Authorized access to the data is managed in partnership with the data providing organizations. The EGA includes major reference data collections for human genetics research.
Collapse
Affiliation(s)
- Ilkka Lappalainen
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Jeff Almeida-King
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Vasudev Kumanduri
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Alexander Senf
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - John Dylan Spalding
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Saif Ur-Rehman
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Gary Saunders
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Jag Kandasamy
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Mario Caccamo
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Rasko Leinonen
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Brendan Vaughan
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Thomas Laurent
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Francis Rowland
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Pablo Marin-Garcia
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Jonathan Barker
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Petteri Jokinen
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | | | | | | | - Ignacio Medina
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | | | | | | | - Arcadi Navarro
- 1] Centre for Genomic Regulation, Barcelona, Spain. [2] Institute of Evolutionary Biology, Universitat Pompeu Fabra-Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain. [3] Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Justin Paschall
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| | - Paul Flicek
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, UK
| |
Collapse
|
382
|
Horikoshi M, Mӓgi R, van de Bunt M, Surakka I, Sarin AP, Mahajan A, Marullo L, Thorleifsson G, Hӓgg S, Hottenga JJ, Ladenvall C, Ried JS, Winkler TW, Willems SM, Pervjakova N, Esko T, Beekman M, Nelson CP, Willenborg C, Wiltshire S, Ferreira T, Fernandez J, Gaulton KJ, Steinthorsdottir V, Hamsten A, Magnusson PKE, Willemsen G, Milaneschi Y, Robertson NR, Groves CJ, Bennett AJ, Lehtimӓki T, Viikari JS, Rung J, Lyssenko V, Perola M, Heid IM, Herder C, Grallert H, Müller-Nurasyid M, Roden M, Hypponen E, Isaacs A, van Leeuwen EM, Karssen LC, Mihailov E, Houwing-Duistermaat JJ, de Craen AJM, Deelen J, Havulinna AS, Blades M, Hengstenberg C, Erdmann J, Schunkert H, Kaprio J, Tobin MD, Samani NJ, Lind L, Salomaa V, Lindgren CM, Slagboom PE, Metspalu A, van Duijn CM, Eriksson JG, Peters A, Gieger C, Jula A, Groop L, Raitakari OT, Power C, Penninx BWJH, de Geus E, Smit JH, Boomsma DI, Pedersen NL, Ingelsson E, Thorsteinsdottir U, Stefansson K, Ripatti S, Prokopenko I, McCarthy MI, Morris AP, ENGAGE Consortium. Discovery and Fine-Mapping of Glycaemic and Obesity-Related Trait Loci Using High-Density Imputation. PLoS Genet 2015; 11:e1005230. [PMID: 26132169 PMCID: PMC4488845 DOI: 10.1371/journal.pgen.1005230] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/18/2015] [Indexed: 11/19/2022] Open
Abstract
Reference panels from the 1000 Genomes (1000G) Project Consortium provide near complete coverage of common and low-frequency genetic variation with minor allele frequency ≥0.5% across European ancestry populations. Within the European Network for Genetic and Genomic Epidemiology (ENGAGE) Consortium, we have undertaken the first large-scale meta-analysis of genome-wide association studies (GWAS), supplemented by 1000G imputation, for four quantitative glycaemic and obesity-related traits, in up to 87,048 individuals of European ancestry. We identified two loci for body mass index (BMI) at genome-wide significance, and two for fasting glucose (FG), none of which has been previously reported in larger meta-analysis efforts to combine GWAS of European ancestry. Through conditional analysis, we also detected multiple distinct signals of association mapping to established loci for waist-hip ratio adjusted for BMI (RSPO3) and FG (GCK and G6PC2). The index variant for one association signal at the G6PC2 locus is a low-frequency coding allele, H177Y, which has recently been demonstrated to have a functional role in glucose regulation. Fine-mapping analyses revealed that the non-coding variants most likely to drive association signals at established and novel loci were enriched for overlap with enhancer elements, which for FG mapped to promoter and transcription factor binding sites in pancreatic islets, in particular. Our study demonstrates that 1000G imputation and genetic fine-mapping of common and low-frequency variant association signals at GWAS loci, integrated with genomic annotation in relevant tissues, can provide insight into the functional and regulatory mechanisms through which their effects on glycaemic and obesity-related traits are mediated. Human genetic studies have demonstrated that quantitative human anthropometric and metabolic traits, including body mass index, waist-hip ratio, and plasma concentrations of glucose and insulin, are highly heritable, and are established risk factors for type 2 diabetes and cardiovascular diseases. Although many regions of the genome have been associated with these traits, the specific genes responsible have not yet been identified. By making use of advanced statistical “imputation” techniques applied to more than 87,000 individuals of European ancestry, and publicly available “reference panels” of more than 37 million genetic variants, we have been able to identify novel regions of the genome associated with these glycaemic and obesity-related traits and localise genes within these regions that are most likely to be causal. This improved understanding of the biological mechanisms underlying glycaemic and obesity-related traits is extremely important because it may advance drug development for downstream disease endpoints, ultimately leading to public health benefits.
Collapse
Affiliation(s)
- Momoko Horikoshi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | - Reedik Mӓgi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Martijn van de Bunt
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Ida Surakka
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Letizia Marullo
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | - Sara Hӓgg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Sciences, Molecular Epidemiology, and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University Amsterdam, Amsterdam, The Netherlands
| | - Claes Ladenvall
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Sara M. Willems
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Children’s Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marian Beekman
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research Leicester Cardiovascular Disease Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Christina Willenborg
- Institute for Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Steven Wiltshire
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Juan Fernandez
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Kyle J. Gaulton
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Anders Hamsten
- Cardiovascular Genetics and Genomics Group, Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Patrik K. E. Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University Amsterdam, Amsterdam, The Netherlands
| | - Yuri Milaneschi
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Neil R. Robertson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Christopher J. Groves
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Amanda J. Bennett
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Terho Lehtimӓki
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| | - Jorma S. Viikari
- Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland
| | - Johan Rung
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
- Steno Diabetes Center A/S, Gentofte, Denmark
| | - Markus Perola
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Germany
| | - Harald Grallert
- 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
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Elina Hypponen
- School of Population Health, University of South Australia, Adelaide, Australia
- Centre for Paediatric Epidemiology and Biostatistics, University College London Institute of Child Health, London, United Kingdom
| | - Aaron Isaacs
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Elisabeth M. van Leeuwen
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lennart C. Karssen
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | - Anton J. M. de Craen
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Joris Deelen
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - Aki S. Havulinna
- Unit of Chronic Disease Epidemiology and Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Matthew Blades
- Bioinformatics and Biostatistics Support Hub (B/BASH), University of Leicester, Leicester, United Kingdom
| | - Christian Hengstenberg
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Munich, Munich, Germany
| | - Jeanette Erdmann
- Institute for Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Munich, Munich, Germany
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- The Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research Leicester Cardiovascular Disease Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Akademiska Sjukhuset, Uppsala, Sweden
| | - Veikko Salomaa
- Unit of Chronic Disease Epidemiology and Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Cecilia M. Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| | - P. Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Johan G. Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Center, Helsinki, Finland
- Vasa Central Hospital, Vasa, Finland
- Department of Health Promotion and Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Annette Peters
- 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
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- 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
| | - Antti Jula
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland
| | - Leif Groop
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Olli T. Raitakari
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Chris Power
- Centre for Paediatric Epidemiology and Biostatistics, University College London Institute of Child Health, London, United Kingdom
| | | | - Eco de Geus
- Department of Biological Psychology, VU University Amsterdam, Amsterdam, The Netherlands
- EMGO Institute for Health and Care Research, VU University & VU University Medical Center, Amsterdam, The Netherlands
| | - Johannes H. Smit
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University Amsterdam, Amsterdam, The Netherlands
| | - Nancy L. Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Erik Ingelsson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Medical Sciences, Molecular Epidemiology, and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Unnur Thorsteinsdottir
- deCode Genetic - Amgen Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCode Genetic - Amgen Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- The Department of Public Health, University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Inga Prokopenko
- Deparment of Genomics of Common Disease, School of Public Health, Imperial College London, London, United Kingdom
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Department of Biostatistics, University of Liverpool, Liverpool, United Kingdom
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
383
|
Rada-Iglesias A. Genetic variation within transcriptional regulatory elements and its implications for human disease. Biol Chem 2015; 395:1453-60. [PMID: 25205712 DOI: 10.1515/hsz-2014-0109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/28/2014] [Indexed: 01/08/2023]
Abstract
Common human pathologies have a complicated etiology involving both genetic and environmental risk factors. Moreover, the genetic basis of these disorders is also complex, with multiple and weak genetic variants contributing to disease susceptibility. In addition, most of these risk genetic variants occur outside genes, within the vast non-coding human genomic space. In this review I first illustrate how large-scale genomic studies aimed at mapping cis-regulatory elements in the human genome are facilitating the identification of disease-causative non-coding genetic variation. I then discuss some of the challenges that remain to be solved before the pathological consequences of non-coding genetic variation can be fully appreciated. Ultimately, revealing the genetics of human complex disease can be a critical step towards more personalized and effective diagnosis and treatments.
Collapse
|
384
|
Structural forms of the human amylase locus and their relationships to SNPs, haplotypes and obesity. Nat Genet 2015; 47:921-5. [PMID: 26098870 PMCID: PMC4712930 DOI: 10.1038/ng.3340] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/27/2015] [Indexed: 12/16/2022]
Abstract
Hundreds of genes reside in structurally complex, poorly understood regions of the human genome1-3. One such region contains the three amylase genes (AMY2B, AMY2A, and AMY1) responsible for digesting starch into sugar. The copy number of AMY1 is reported to be the genome’s largest influence on obesity4, though genome-wide association studies for obesity have found this locus unremarkable. Using whole genome sequence analysis3,5, droplet digital PCR6, and genome mapping7, we identified eight common structural haplotypes of the amylase locus that suggest its mutational history. We found that AMY1 copy number in individuals’ genomes is generally even (rather than odd) and partially correlates to nearby SNPs, which do not associate with BMI. We measured amylase gene copy number in 1,000 obese or lean Estonians and in two other cohorts totaling ~3,500 individuals. We had 99% power to detect the lower bound of the reported effects on BMI4, yet found no association.
Collapse
|
385
|
Vasques GA, Arnhold IJP, Jorge AAL. Role of the natriuretic peptide system in normal growth and growth disorders. Horm Res Paediatr 2015; 82:222-9. [PMID: 25196103 DOI: 10.1159/000365049] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/03/2014] [Indexed: 11/19/2022] Open
Abstract
The C-type natriuretic peptide (CNP) and its receptor (NPR-B) are recognized as important regulators of longitudinal growth. Animal models involving CNP or NPR-B genes (Nppc or Npr2) support the fundamental role of CNP/NPR-B for endochondral ossification. Studies with these animals allow the development of potential drug therapies for dwarfism. Polymorphisms in two genes related to the CNP pathway have been implicated in height variability in healthy individuals. Biallelic loss-of-function mutations in NPR-B gene (NPR2) cause acromesomelic dysplasia type Maroteux, a skeletal dysplasia with extremely short stature. Heterozygous mutations in NPR2 are responsible for nonsyndromic familial short stature. Conversely, heterozygous gain-of-function mutations in NPR2 cause tall stature, with a variable phenotype. A phase 2 multicenter and multinational trial is being developed to evaluate a CNP analog treatment for achondroplasia. Pediatricians and endocrinologists must be aware of growth disorders related to natriuretic peptides, although there is still much to be learned about its diagnostic and therapeutic use.
Collapse
Affiliation(s)
- Gabriela A Vasques
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM-25, Universidade de São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
386
|
Genome-wide association study identifies novel genetic variants contributing to variation in blood metabolite levels. Nat Commun 2015; 6:7208. [PMID: 26068415 DOI: 10.1038/ncomms8208] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 04/20/2015] [Indexed: 01/06/2023] Open
Abstract
Metabolites are small molecules involved in cellular metabolism, which can be detected in biological samples using metabolomic techniques. Here we present the results of genome-wide association and meta-analyses for variation in the blood serum levels of 129 metabolites as measured by the Biocrates metabolomic platform. In a discovery sample of 7,478 individuals of European descent, we find 4,068 genome- and metabolome-wide significant (Z-test, P < 1.09 × 10(-9)) associations between single-nucleotide polymorphisms (SNPs) and metabolites, involving 59 independent SNPs and 85 metabolites. Five of the fifty-nine independent SNPs are new for serum metabolite levels, and were followed-up for replication in an independent sample (N = 1,182). The novel SNPs are located in or near genes encoding metabolite transporter proteins or enzymes (SLC22A16, ARG1, AGPS and ACSL1) that have demonstrated biomedical or pharmaceutical importance. The further characterization of genetic influences on metabolic phenotypes is important for progress in biological and medical research.
Collapse
|
387
|
Luna-Luna M, Medina-Urrutia A, Vargas-Alarcón G, Coss-Rovirosa F, Vargas-Barrón J, Pérez-Méndez Ó. Adipose Tissue in Metabolic Syndrome: Onset and Progression of Atherosclerosis. Arch Med Res 2015; 46:392-407. [PMID: 26009250 DOI: 10.1016/j.arcmed.2015.05.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/12/2015] [Indexed: 12/25/2022]
Abstract
Metabolic syndrome (MetS) should be considered a clinical entity when its different symptoms share a common etiology: obesity/insulin resistance as a result of a multi-organ dysfunction. The main interest in treating MetS as a clinical entity is that the addition of its components drastically increases the risk of atherosclerosis. In MetS, the adipose tissue plays a central role along with an unbalanced gut microbiome, which has become relevant in recent years. Once visceral adipose tissue (VAT) increases, dyslipidemia and endothelial dysfunction follow as additive risk factors. However, when the nonalcoholic fatty liver is present, risk of a cardiovascular event is highly augmented. Epicardial adipose tissue (EAT) seems to increase simultaneously with the VAT. In this context, the former may play a more important role in the development of the atherosclerotic plaque than the latter. Hence, EAT may act as a paracrine tissue vis-à-vis the coronary arteries favoring the local inflammation and the atheroma calcification.
Collapse
Affiliation(s)
- María Luna-Luna
- Department of Molecular Biology, Instituto Nacional de Cardiología, Mexico City, Mexico
| | | | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, Instituto Nacional de Cardiología, Mexico City, Mexico; Study Group of Atherosclerosis, Instituto Nacional de Cardiología, Mexico City, Mexico
| | | | - Jesús Vargas-Barrón
- Echocardiography, Instituto Nacional de Cardiología, Mexico City, Mexico; Study Group of Atherosclerosis, Instituto Nacional de Cardiología, Mexico City, Mexico
| | - Óscar Pérez-Méndez
- Department of Molecular Biology, Instituto Nacional de Cardiología, Mexico City, Mexico; Study Group of Atherosclerosis, Instituto Nacional de Cardiología, Mexico City, Mexico.
| |
Collapse
|
388
|
Potential Signal Transduction Regulation by HDL of the β2-Adrenergic Receptor Pathway. Implications in Selected Pathological Situations. Arch Med Res 2015; 46:361-71. [PMID: 26009249 DOI: 10.1016/j.arcmed.2015.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/12/2015] [Indexed: 01/09/2023]
Abstract
The main atheroprotective mechanism of high-density lipoprotein (HDL) has been regarded as reverse cholesterol transport, whereby cholesterol from peripheral tissues is removed and transported to the liver for elimination. Although numerous additional atheroprotective mechanisms have been suggested, the role of HDL in modulating signal transduction of cell membrane-bound receptors has received little attention to date. This potential was recently highlighted following the identification of a polymorphism in the adenylyl cyclase 9 gene (ADCY9) that was shown to be a determining factor in the risk of cardiovascular (CV) events in patients treated with the HDL-raising compound dalcetrapib. Indeed, ADCY9 is part of the signaling pathway of the β2-adrenergic receptor (β2-AR) and both are membrane-bound proteins affected by changes in membrane-rich cholesterol plasma membrane domains (caveolae). Numerous G-protein-coupled receptors (GPCRs) and ion channels are affected by caveolae, with caveolae composition acting as a 'signalosome'. Polymorphisms in the genes encoding ADCY9 and β2-AR are associated with response to β2-agonist drugs in patients with asthma, malaria and with sickle cell disease. Crystallization of the β2-AR has found cholesterol tightly bound to transmembrane structures of the receptor. Cholesterol has also been shown to modulate the activity of this receptor. Apolipoprotein A1 (ApoA1), the major protein component of HDL, destabilizes and removes cholesterol from caveolae with high affinity through interaction with ATP-binding cassette transporter. Furthermore, β2-AR activity may be affected by ApoA1/HDL-targeted therapies. Taken together, these observations suggest a common pathway that potentially links a primary HDL function to the regulation of signal transduction.
Collapse
|
389
|
Mariman ECM, Szklarczyk R, Bouwman FG, Aller EEJG, van Baak MA, Wang P. Olfactory receptor genes cooperate with protocadherin genes in human extreme obesity. GENES AND NUTRITION 2015; 10:465. [PMID: 25943692 PMCID: PMC4420755 DOI: 10.1007/s12263-015-0465-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/21/2015] [Indexed: 01/28/2023]
Abstract
Worldwide, the incidence of obesity has increased dramatically over the past decades. More knowledge about the complex etiology of obesity is needed in order to find additional approaches for treatment and prevention. Investigating the exome sequencing data of 30 extremely obese subjects (BMI 45-65 kg/m(2)) shows that predicted damaging missense variants in olfactory receptor genes on chromosome 1q and rare predicted damaging variants in the protocadherin (PCDH) beta-cluster genes on chromosome 5q31, reported in our previous work, co-localize in subjects with extreme obesity. This implies a synergistic effect between genetic variation in these gene clusters in the predisposition to extreme obesity. Evidence for a general involvement of the olfactory transduction pathway on itself could not be found. Bioinformatic analysis indicates a specific involvement of the PCDH beta-cluster genes in controlling tissue development. Further mechanistic insight needs to await the identification of the ligands of the 1q olfactory receptors. Eventually, this may provide the possibility to manipulate food flavor in a way to reduce the risk of overeating and of extreme obesity in genetically predisposed subjects.
Collapse
Affiliation(s)
- Edwin C M Mariman
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands,
| | | | | | | | | | | |
Collapse
|
390
|
Sun L, Deng H, He L, Hu X, Huang Q, Xue J, Chen J, Shi X, Xu Y. The relationship between NR2E1 and subclinical inflammation in newly diagnosed type 2 diabetic patients. J Diabetes Complications 2015; 29:589-94. [PMID: 25813674 DOI: 10.1016/j.jdiacomp.2014.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/25/2014] [Accepted: 12/26/2014] [Indexed: 02/06/2023]
Abstract
AIMS To evaluate the expression level of NR2E1 (nuclear receptor subfamily 2,group E,member 1) and its correlation with type 2 diabetes (T2DM). METHODS Plasma and peripheral blood mononuclear cells (PBMCs) were collected from 54 T2DM and 88 healthy individuals. The levels of free fatty acids (FFAs), total cholesterol (TC), triglyceride (TG), high density lipoprotein (HDL-c), low density lipoprotein (LDL-c), fasting insulin (FIN), and fasting blood glucose (FBG) were measured. The insulin resistance index was calculated using the homeostasis model assessment (HOMA). NR2E1 in PBMCs were analyzed using real-time RT-PCR and Western blots. Tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) in the plasma were measured by enzyme-linked immunosorbent assay (ELISA). PBMCs isolated from healthy volunteers were treated with glucose or palmitate for 24h, followed by analysis for the expression level of NR2E1.The amount of TNF-α and IL-6 secreted into the supernatant were measured by ELISA. RESULTS FIN, FBG, HOMA and TNF-α, IL-6 were significantly higher in diabetic patients, compared to the control group. Levels of NR2E1 were significantly higher in the PBMCs isolated from the diabetic group, compared to the control group. NR2E1 expression was positively correlated with FBG, FIN, HOMA, FFAs, TNF-α and IL-6. Glucose and palmitate treatment significantly increased NR2E1 gene expression and inflammatory cytokines production in PBMCs in vitro. CONCLUSIONS Increased NR2E1 level may be closely associated with inflammation and disorder of lipid and glucose metabolism in diabetic patients.
Collapse
Affiliation(s)
- Li Sun
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Haohua Deng
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Lanjie He
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Xuemei Hu
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Qi Huang
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Junli Xue
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Jin Chen
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Xiaoli Shi
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Yancheng Xu
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China.
| |
Collapse
|
391
|
Abstract
Genome-wide association studies that scan the genome for common genetic variants associated with phenotype have greatly advanced medical knowledge. Hyperuricemia is no exception, with 28 loci identified. However, genetic control of pathways determining gout in the presence of hyperuricemia is still poorly understood. Two important pathways determining hyperuricemia have been confirmed (renal and gut excretion of uric acid with glycolysis now firmly implicated). Major urate loci are SLC2A9 and ABCG2. Recent studies show that SLC2A9 is involved in renal and gut excretion of uric acid and is implicated in antioxidant defense. Although etiological variants at SLC2A9 are yet to be identified, it is clear that considerable genetic complexity exists at the SLC2A9 locus, with multiple statistically independent genetic variants and local epistatic interactions. The positions of implicated genetic variants within or near chromatin regions involved in transcriptional control suggest that this mechanism (rather than structural changes in SLC2A9) is important in regulating the activity of SLC2A9. ABCG2 is involved primarily in extra-renal uric acid under-excretion with the etiological variant influencing expression. At the other 26 loci, probable causal genes can be identified at three (PDZK1, SLC22A11, and INHBB) with strong candidates at a further 10 loci. Confirmation of the causal gene will require a combination of re-sequencing, trans-ancestral mapping, and correlation of genetic association data with expression data. As expected, the urate loci associate with gout, although inconsistent effect sizes for gout require investigation. Finally, there has been no genome-wide association study using clinically ascertained cases to investigate the causes of gout in the presence of hyperuricemia. In such a study, use of asymptomatic hyperurcemic controls would be expected to increase the ability to detect genetic associations with gout.
Collapse
Affiliation(s)
- Tony R Merriman
- Department of Biochemistry, University of Otago, Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
392
|
Paré G, Asma S, Deng WQ. Contribution of large region joint associations to complex traits genetics. PLoS Genet 2015; 11:e1005103. [PMID: 25856144 PMCID: PMC4391841 DOI: 10.1371/journal.pgen.1005103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 02/26/2015] [Indexed: 02/03/2023] Open
Abstract
A polygenic model of inheritance, whereby hundreds or thousands of weakly associated variants contribute to a trait’s heritability, has been proposed to underlie the genetic architecture of complex traits. However, relatively few genetic variants have been positively identified so far and they collectively explain only a small fraction of the predicted heritability. We hypothesized that joint association of multiple weakly associated variants over large chromosomal regions contributes to complex traits variance. Confirmation of such regional associations can help identify new loci and lead to a better understanding of known ones. To test this hypothesis, we first characterized the ability of commonly used genetic association models to identify large region joint associations. Through theoretical derivation and simulation, we showed that multivariate linear models where multiple SNPs are included as independent predictors have the most favorable association profile. Based on these results, we tested for large region association with height in 3,740 European participants from the Health and Retirement Study (HRS) study. Adjusting for SNPs with known association with height, we demonstrated clustering of weak associations (p = 2x10-4) in regions extending up to 433.0 Kb from known height loci. The contribution of regional associations to phenotypic variance was estimated at 0.172 (95% CI 0.063-0.279; p < 0.001), which compared favorably to 0.129 explained by known height variants. Conversely, we showed that suggestively associated regions are enriched for known height loci. To extend our findings to other traits, we also tested BMI, HDLc and CRP for large region associations, with consistent results for CRP. Our results demonstrate the presence of large region joint associations and suggest these can be used to pinpoint weakly associated SNPs. It is widely accepted that genetics influences a broad range of human traits and diseases, yet only a few genetic variants are known to determine these traits and their impact is modest. In this report, we made the hypothesis that combining information from a large number of genetic variants would help better explain how they together contribute to traits such as height. To do so, we first had to select a proper method to integrate large numbers of genetic variants in a single test, here named “large region joint association”. Next, we tested our method on height in 3,740 European participants from the Health and Retirement Study. We showed that the contribution of regional associations to variation in height was 17.2%, as compared to the 12.9% explained by known genetic determinants of height. In other words, the joint effect of multiple genetic variants integrated together contributed to a substantial fraction of the genetics of height. These results are significant because they can help identify new genes or genetic regions associated with human traits or diseases. Conversely, these results can be used to better understand genes that we already know are associated. Furthermore, our results provide insights on how traits are genetically determined.
Collapse
Affiliation(s)
- Guillaume Paré
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- Population Genomics Program, Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Canada
- * E-mail:
| | - Senay Asma
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Canada
| | - Wei Q. Deng
- Department of Statistical Sciences, University of Toronto, Toronto, Canada
| |
Collapse
|
393
|
Ollikainen M, Ismail K, Gervin K, Kyllönen A, Hakkarainen A, Lundbom J, Järvinen EA, Harris JR, Lundbom N, Rissanen A, Lyle R, Pietiläinen KH, Kaprio J. Genome-wide blood DNA methylation alterations at regulatory elements and heterochromatic regions in monozygotic twins discordant for obesity and liver fat. Clin Epigenetics 2015; 7:39. [PMID: 25866590 PMCID: PMC4393626 DOI: 10.1186/s13148-015-0073-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/11/2015] [Indexed: 12/16/2022] Open
Abstract
Background The current epidemic of obesity and associated diseases calls for swift actions to better understand the mechanisms by which genetics and environmental factors affect metabolic health in humans. Monozygotic (MZ) twin pairs showing discordance for obesity suggest that epigenetic influences represent one such mechanism. We studied genome-wide leukocyte DNA methylation variation in 30 clinically healthy young adult MZ twin pairs discordant for body mass index (BMI; average within-pair BMI difference: 5.4 ± 2.0 kg/m2). Results There were no differentially methylated cytosine-guanine (CpG) sites between the co-twins discordant for BMI. However, stratification of the twin pairs based on the level of liver fat accumulation revealed two epigenetically highly different groups. Significant DNA methylation differences (n = 1,236 CpG sites (CpGs)) between the co-twins were only observed if the heavier co-twins had excessive liver fat (n = 13 twin pairs). This unhealthy pattern of obesity was coupled with insulin resistance and low-grade inflammation. The differentially methylated CpGs included 23 genes known to be associated with obesity, liver fat, type 2 diabetes mellitus (T2DM) and metabolic syndrome, and potential novel metabolic genes. Differentially methylated CpG sites were overrepresented at promoters, insulators, and heterochromatic and repressed regions. Based on predictions by overlapping histone marks, repressed and weakly transcribed sites were significantly more often hypomethylated, whereas sites with strong enhancers and active promoters were hypermethylated. Further, significant clustering of differentially methylated genes in vitamin, amino acid, fatty acid, sulfur, and renin-angiotensin metabolism pathways was observed. Conclusions The methylome in leukocytes is altered in obesity associated with metabolic disturbances, and our findings indicate several novel candidate genes and pathways in obesity and obesity-related complications. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0073-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miina Ollikainen
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Khadeeja Ismail
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Kristina Gervin
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Anjuska Kyllönen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Jesper Lundbom
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Elina A Järvinen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Jennifer R Harris
- Division of Epidemiology, The Norwegian Institute of Public Health, Oslo, Norway
| | - Nina Lundbom
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Aila Rissanen
- Department of Psychiatry, Helsinki University Central Hospital, Helsinki, Finland.,Endocrinology, Abdominal Center, Helsinki University Central Hospital, Helsinki, Finland
| | - Robert Lyle
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Endocrinology, Abdominal Center, Helsinki University Central Hospital, Helsinki, Finland.,Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, Helsinki, Finland.,Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland.,Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
394
|
Abstract
The rising global prevalence of diabetes mellitus is accompanied by an increasing burden of morbidity and mortality that is attributable to the complications of chronic hyperglycaemia. These complications include blindness, renal failure and cardiovascular disease. Current therapeutic options for chronic hyperglycaemia reduce, but do not eradicate, the risk of these complications. Success in defining new preventative and therapeutic strategies hinges on an improved understanding of the molecular processes involved in the development of these complications. This Review explores the role of human genetics in delivering such insights, and describes progress in characterizing the sequence variants that influence individual predisposition to diabetic kidney disease, retinopathy, neuropathy and accelerated cardiovascular disease. Numerous risk variants for microvascular complications of diabetes have been reported, but very few have shown robust replication. Furthermore, only limited evidence exists of a difference in the repertoire of risk variants influencing macrovascular disease between those with and those without diabetes. Here, we outline the challenges associated with the genetic analysis of diabetic complications and highlight ongoing efforts to deliver biological insights that can drive translational benefits.
Collapse
|
395
|
Xu Z, Duan Q, Yan S, Chen W, Li M, Lange E, Li Y. DISSCO: direct imputation of summary statistics allowing covariates. Bioinformatics 2015; 31:2434-42. [PMID: 25810429 DOI: 10.1093/bioinformatics/btv168] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 03/17/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Imputation of individual level genotypes at untyped markers using an external reference panel of genotyped or sequenced individuals has become standard practice in genetic association studies. Direct imputation of summary statistics can also be valuable, for example in meta-analyses where individual level genotype data are not available. Two methods (DIST and ImpG-Summary/LD), that assume a multivariate Gaussian distribution for the association summary statistics, have been proposed for imputing association summary statistics. However, both methods assume that the correlations between association summary statistics are the same as the correlations between the corresponding genotypes. This assumption can be violated in the presence of confounding covariates. METHODS We analytically show that in the absence of covariates, correlation among association summary statistics is indeed the same as that among the corresponding genotypes, thus serving as a theoretical justification for the recently proposed methods. We continue to prove that in the presence of covariates, correlation among association summary statistics becomes the partial correlation of the corresponding genotypes controlling for covariates. We therefore develop direct imputation of summary statistics allowing covariates (DISSCO). RESULTS We consider two real-life scenarios where the correlation and partial correlation likely make practical difference: (i) association studies in admixed populations; (ii) association studies in presence of other confounding covariate(s). Application of DISSCO to real datasets under both scenarios shows at least comparable, if not better, performance compared with existing correlation-based methods, particularly for lower frequency variants. For example, DISSCO can reduce the absolute deviation from the truth by 3.9-15.2% for variants with minor allele frequency <5%.
Collapse
Affiliation(s)
- Zheng Xu
- Department of Biostatistics, Department of Genetics, Department of Computer Science
| | - Qing Duan
- Department of Genetics, Curriculum in Bioinformatics and Computational Biology, Department of Statistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Song Yan
- Department of Biostatistics, Department of Genetics, Department of Computer Science
| | - Wei Chen
- Division of Pediatric Pulmonary Medicine, Allergy and Immunology, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Department of Biostatistics, Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA 15224, USA and
| | - Mingyao Li
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ethan Lange
- Department of Biostatistics, Department of Genetics
| | - Yun Li
- Department of Biostatistics, Department of Genetics, Department of Computer Science
| |
Collapse
|
396
|
Yazdi FT, Clee SM, Meyre D. Obesity genetics in mouse and human: back and forth, and back again. PeerJ 2015; 3:e856. [PMID: 25825681 PMCID: PMC4375971 DOI: 10.7717/peerj.856] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 03/05/2015] [Indexed: 12/19/2022] Open
Abstract
Obesity is a major public health concern. This condition results from a constant and complex interplay between predisposing genes and environmental stimuli. Current attempts to manage obesity have been moderately effective and a better understanding of the etiology of obesity is required for the development of more successful and personalized prevention and treatment options. To that effect, mouse models have been an essential tool in expanding our understanding of obesity, due to the availability of their complete genome sequence, genetically identified and defined strains, various tools for genetic manipulation and the accessibility of target tissues for obesity that are not easily attainable from humans. Our knowledge of monogenic obesity in humans greatly benefited from the mouse obesity genetics field. Genes underlying highly penetrant forms of monogenic obesity are part of the leptin-melanocortin pathway in the hypothalamus. Recently, hypothesis-generating genome-wide association studies for polygenic obesity traits in humans have led to the identification of 119 common gene variants with modest effect, most of them having an unknown function. These discoveries have led to novel animal models and have illuminated new biologic pathways. Integrated mouse-human genetic approaches have firmly established new obesity candidate genes. Innovative strategies recently developed by scientists are described in this review to accelerate the identification of causal genes and deepen our understanding of obesity etiology. An exhaustive dissection of the molecular roots of obesity may ultimately help to tackle the growing obesity epidemic worldwide.
Collapse
Affiliation(s)
- Fereshteh T. Yazdi
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Susanne M. Clee
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - David Meyre
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
397
|
Martínez-Barquero V, de Marco G, Martínez-Hervas S, Rentero P, Galan-Chilet I, Blesa S, Morchon D, Morcillo S, Rojo G, Ascaso JF, Real JT, Martín-Escudero JC, Chaves FJ. Polymorphisms in endothelin system genes, arsenic levels and obesity risk. PLoS One 2015; 10:e0118471. [PMID: 25799405 PMCID: PMC4370725 DOI: 10.1371/journal.pone.0118471] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/18/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND/OBJECTIVES Obesity has been linked to morbidity and mortality through increased risk for many chronic diseases. Endothelin (EDN) system has been related to endothelial function but it can be involved in lipid metabolism regulation: Receptor type A (EDNRA) activates lipolysis in adipocytes, the two endothelin receptors mediate arsenic-stimulated adipocyte dysfunction, and endothelin system can regulate adiposity by modulating adiponectin activity in different situations and, therefore, influence obesity development. The aim of the present study was to analyze if single nucleotide polymorphisms (SNPs) in the EDN system could be associated with human obesity. SUBJECTS/METHODS We analyzed two samples of general-population-based studies from two different regions of Spain: the VALCAR Study, 468 subjects from the area of Valencia, and the Hortega Study, 1502 subjects from the area of Valladolid. Eighteen SNPs throughout five genes were analyzed using SNPlex. RESULTS We found associations for two polymorphisms of the EDNRB gene which codifies for EDN receptor type B. Genotypes AG and AA of the rs5351 were associated with a lower risk for obesity in the VALCAR sample (p=0.048, OR=0.63) and in the Hortega sample (p=0.001, OR=0.62). Moreover, in the rs3759475 polymorphism, genotypes CT and TT were also associated with lower risk for obesity in the Hortega sample (p=0.0037, OR=0.66) and in the VALCAR sample we found the same tendency (p=0.12, OR=0.70). Furthermore, upon studying the pooled population, we found a stronger association with obesity (p=0.0001, OR=0.61 and p=0.0008, OR=0.66 for rs5351 and rs3759475, respectively). Regarding plasma arsenic levels, we have found a positive association for the two SNPs studied with obesity risk in individuals with higher arsenic levels in plasma: rs5351 (p=0.0054, OR=0.51) and rs3759475 (p=0.009, OR=0.53). CONCLUSIONS Our results support the hypothesis that polymorphisms of the EDNRB gene may influence the susceptibility to obesity and can interact with plasma arsenic levels.
Collapse
Affiliation(s)
- Vanesa Martínez-Barquero
- Department of Medicine, University of Valencia, Valencia, Spain
- Genotyping and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Griselda de Marco
- Genotyping and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Sergio Martínez-Hervas
- Department of Medicine, University of Valencia, Valencia, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
- Service of Endocrinology and Nutrition, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Pilar Rentero
- Genotyping and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Inmaculada Galan-Chilet
- Genotyping and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Sebastian Blesa
- Genotyping and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - David Morchon
- Internal Medicine, Rio Hortega Hospital, University of Valladolid, Valladolid, Spain
| | - Sonsoles Morcillo
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
- Service of Endocrinology and Nutrition, Hospital Regional Universitario, Málaga, Spain, Instituto de Biomedicina de Málaga (IBIMA), Málaga, Spain
| | - Gemma Rojo
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
- Service of Endocrinology and Nutrition, Hospital Regional Universitario, Málaga, Spain, Instituto de Biomedicina de Málaga (IBIMA), Málaga, Spain
| | - Juan Francisco Ascaso
- Department of Medicine, University of Valencia, Valencia, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
- Service of Endocrinology and Nutrition, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - José Tomás Real
- Department of Medicine, University of Valencia, Valencia, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
- Service of Endocrinology and Nutrition, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | - Felipe Javier Chaves
- Genotyping and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
- * E-mail:
| |
Collapse
|
398
|
Liu X, Hinney A, Scholz M, Scherag A, Tönjes A, Stumvoll M, Stadler PF, Hebebrand J, Böttcher Y. Indications for potential parent-of-origin effects within the FTO gene. PLoS One 2015; 10:e0119206. [PMID: 25793382 PMCID: PMC4368796 DOI: 10.1371/journal.pone.0119206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 01/28/2015] [Indexed: 12/18/2022] Open
Abstract
Genome-Wide Association Studies (GWAS) were successfully applied to discover associations with obesity. However, the GWAS design is usually based on unrelated individuals and inheritance information on the parental origin of the alleles is missing. Taking into account parent-of-origin may provide further insights into the genetic mechanisms contributing to obesity. We hypothesized that there may be variants within the robustly replicated fat mass and obesity associated (FTO) gene that may confer different risk for obesity depending on transmission from mother or father. Genome-wide genotypes and pedigree information from the Sorbs population were used. Phased genotypes among 525 individuals were generated by AlphaImpute. Subsequently, 22 SNPs within FTO introns 1 to 3 were selected and parent-of-origin specific association analyses were performed using PLINK. Interestingly, we identified several SNPs conferring different genetic effects (P≤0.05) depending on parental origin—among them, rs1861868, rs1121980 and rs9939973 (all in intron 1). To confirm our findings, we investigated the selected variants in 705 German trios comprising an (extremely) obese child or adolescent and both parents. Again, we observed evidence for POE effects in intron 2 and 3 (P≤0.05) as indicated by the parental asymmetry test. Our results suggest that the obesity risk transmitted by several FTO variants may depend on the parental origin of the allele. Larger family-based studies are warranted to replicate our findings.
Collapse
Affiliation(s)
- Xuanshi Liu
- IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Bioinformatics Group, Department of Computer Science, University of Leipzig, Leipzig, Germany
| | - Anke Hinney
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy Universitätsklinikum Essen, University of Duisburg-Essen, Essen, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - André Scherag
- Clinical Epidemiology, Integrated Research and Treatment Center (IFB) Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Anke Tönjes
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Peter F. Stadler
- Fraunhofer Institute for Cell Therapy and Immunology, AG RNomics, Leipzig, Germany
- Interdisciplinary Center of Bioinformatics, University of Leipzig, Leipzig, Germany
- Institute for Theoretical Chemistry, University of Vienna, Vienna, Austria
- Sante Fe Institute, Santa Fe, New Mexico, United States of America
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy Universitätsklinikum Essen, University of Duisburg-Essen, Essen, Germany
| | - Yvonne Böttcher
- IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- * E-mail: (YB)
| |
Collapse
|
399
|
Nead KT, Li A, Wehner MR, Neupane B, Gustafsson S, Butterworth A, Engert JC, Davis AD, Hegele RA, Miller R, den Hoed M, Khaw KT, Kilpeläinen TO, Wareham N, Edwards TL, Hallmans G, Varga TV, Kardia SLR, Smith JA, Zhao W, Faul JD, Weir D, Mi J, Xi B, Quinteros SC, Cooper C, Sayer AA, Jameson K, Grøntved A, Fornage M, Sidney S, Hanis CL, Highland HM, Häring HU, Heni M, Lasky-Su J, Weiss ST, Gerhard GS, Still C, Melka MM, Pausova Z, Paus T, Grant SFA, Hakonarson H, Price RA, Wang K, Scherag A, Hebebrand J, Hinney A, Franks PW, Frayling TM, McCarthy MI, Hirschhorn JN, Loos RJ, Ingelsson E, Gerstein HC, Yusuf S, Beyene J, Anand SS, Meyre D. Contribution of common non-synonymous variants in PCSK1 to body mass index variation and risk of obesity: a systematic review and meta-analysis with evidence from up to 331 175 individuals. Hum Mol Genet 2015; 24:3582-94. [PMID: 25784503 DOI: 10.1093/hmg/ddv097] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/13/2015] [Indexed: 12/31/2022] Open
Abstract
Polymorphisms rs6232 and rs6234/rs6235 in PCSK1 have been associated with extreme obesity [e.g. body mass index (BMI) ≥ 40 kg/m(2)], but their contribution to common obesity (BMI ≥ 30 kg/m(2)) and BMI variation in a multi-ethnic context is unclear. To fill this gap, we collected phenotypic and genetic data in up to 331 175 individuals from diverse ethnic groups. This process involved a systematic review of the literature in PubMed, Web of Science, Embase and the NIH GWAS catalog complemented by data extraction from pre-existing GWAS or custom-arrays in consortia and single studies. We employed recently developed global meta-analytic random-effects methods to calculate summary odds ratios (OR) and 95% confidence intervals (CIs) or beta estimates and standard errors (SE) for the obesity status and BMI analyses, respectively. Significant associations were found with binary obesity status for rs6232 (OR = 1.15, 95% CI 1.06-1.24, P = 6.08 × 10(-6)) and rs6234/rs6235 (OR = 1.07, 95% CI 1.04-1.10, P = 3.00 × 10(-7)). Similarly, significant associations were found with continuous BMI for rs6232 (β = 0.03, 95% CI 0.00-0.07; P = 0.047) and rs6234/rs6235 (β = 0.02, 95% CI 0.00-0.03; P = 5.57 × 10(-4)). Ethnicity, age and study ascertainment significantly modulated the association of PCSK1 polymorphisms with obesity. In summary, we demonstrate evidence that common gene variation in PCSK1 contributes to BMI variation and susceptibility to common obesity in the largest known meta-analysis published to date in genetic epidemiology.
Collapse
Affiliation(s)
- Kevin T Nead
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Aihua Li
- Department of Clinical Epidemiology and Biostatistics
| | - Mackenzie R Wehner
- Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Binod Neupane
- Department of Clinical Epidemiology and Biostatistics
| | - Stefan Gustafsson
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Adam Butterworth
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - James C Engert
- Population Health Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton General Hospital, Hamilton, ON, Canada L8L 2X
| | | | - Robert A Hegele
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada L8S 4L8, Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala SE 751 05, Sweden
| | | | - Marcel den Hoed
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada H3H 2R9, Six Nations Health Services, Ohsweken, Canada N0A 1M0
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Tuomas O Kilpeläinen
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada H3H 2R9, Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, London, ON, Canada N6A 5K8
| | - Nick Wareham
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada H3H 2R9
| | - Todd L Edwards
- Department of Medicine, University of Western Ontario, London, ON, Canada N6A 3K7
| | - Göran Hallmans
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Tibor V Varga
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sharon L R Kardia
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jennifer A Smith
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen 2100, Denmark
| | - Wei Zhao
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jessica D Faul
- Center for Human Genetics Research, Vanderbilt Epidemiology Center, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - David Weir
- Center for Human Genetics Research, Vanderbilt Epidemiology Center, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - Jie Mi
- Department of Public Health and Clinical Medicine, Umeå University, Umeå 901 87, Sweden
| | - Bo Xi
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö 205 02, Sweden
| | | | - Cyrus Cooper
- Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA, Department of Epidemiology, Capital Institute of Pediatrics, Beijing 100020, China, Department of Maternal and Child Health Care, School of Public Health, Shandong University, Jinan 250100, China
| | - Avan Aihie Sayer
- Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Karen Jameson
- Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Anders Grøntved
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, Universidad Nacional Autónoma de México, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Myriam Fornage
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Stephen Sidney
- National Institute for Health Research Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Craig L Hanis
- National Institute for Health Research Biomedical Research Unit, University of Oxford, Oxford OX3 7LE, UK
| | - Heather M Highland
- National Institute for Health Research Biomedical Research Unit, University of Oxford, Oxford OX3 7LE, UK
| | - Hans-Ulrich Häring
- Department of Sport Science and Clinical Biomechanics, University of Southern Denmark, Odense DK-5230, Denmark, University of Texas Health Science Center at Houston Institute of Molecular Medicine and Division of Epidemiology Human Genetics and Environmental Sciences, School of Public Health, Houston, TX 77030, USA
| | - Martin Heni
- Department of Sport Science and Clinical Biomechanics, University of Southern Denmark, Odense DK-5230, Denmark, University of Texas Health Science Center at Houston Institute of Molecular Medicine and Division of Epidemiology Human Genetics and Environmental Sciences, School of Public Health, Houston, TX 77030, USA
| | - Jessica Lasky-Su
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA 94612, USA, The Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Scott T Weiss
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA 94612, USA, The Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Glenn S Gerhard
- Internal Medicine IV (Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry), University Hospital of Tuebingen, Tübingen 72076, Germany
| | | | - Melkaey M Melka
- The Department of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zdenka Pausova
- The Department of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomáš Paus
- Center for Genomic Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Struan F A Grant
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Department of Pathology and Laboratory Medicine, Pennsylvania State University, Hershey, PA 17033, USA, Geisinger Obesity Institute, Danville, PA 17822, USA
| | - Hakon Hakonarson
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Department of Pathology and Laboratory Medicine, Pennsylvania State University, Hershey, PA 17033, USA, Geisinger Obesity Institute, Danville, PA 17822, USA
| | - R Arlen Price
- The Hospital for Sick Children, Department of Physiology, University of Toronto, Toronto, ON, Canada M5G 1X
| | - Kai Wang
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK, Rotman Research Institute, University of Toronto, Toronto, Canada M6A 2E1
| | - Andre Scherag
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | - Paul W Franks
- Department of Medicine, University of Western Ontario, London, ON, Canada N6A 3K7, MRC Epidemiology Unit, University of Cambridge, Cambridge, UK, Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy M Frayling
- Zilkha Neurogenetic Institute, Department of Psychiatry and Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Mark I McCarthy
- Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena 07740, Germany
| | - Joel N Hirschhorn
- Department of Child and Adolescent Psychiatry, University of Duisburg-Essen, Essen 45141, Germany, Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA, Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter EX2 4TH, UK
| | - Ruth J Loos
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada H3H 2R9, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 9DU, UK
| | - Erik Ingelsson
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Hertzel C Gerstein
- Department of Clinical Epidemiology and Biostatistics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA, Divisions of Genetics and Endocrinology, Children's Hospital, Boston, MA 02115, USA
| | - Salim Yusuf
- Department of Clinical Epidemiology and Biostatistics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA, Divisions of Genetics and Endocrinology, Children's Hospital, Boston, MA 02115, USA
| | - Joseph Beyene
- Department of Clinical Epidemiology and Biostatistics
| | - Sonia S Anand
- Department of Clinical Epidemiology and Biostatistics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA, Divisions of Genetics and Endocrinology, Children's Hospital, Boston, MA 02115, USA
| | - David Meyre
- Department of Clinical Epidemiology and Biostatistics, Divisions of Genetics and Endocrinology, Children's Hospital, Boston, MA 02115, USA, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA,
| |
Collapse
|
400
|
Albuquerque D, Stice E, Rodríguez-López R, Manco L, Nóbrega C. Current review of genetics of human obesity: from molecular mechanisms to an evolutionary perspective. Mol Genet Genomics 2015; 290:1191-221. [DOI: 10.1007/s00438-015-1015-9] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/11/2015] [Indexed: 12/18/2022]
|