251
|
Strawbridge RJ, Ward J, Cullen B, Tunbridge EM, Hartz S, Bierut L, Horton A, Bailey MES, Graham N, Ferguson A, Lyall DM, Mackay D, Pidgeon LM, Cavanagh J, Pell JP, O'Donovan M, Escott-Price V, Harrison PJ, Smith DJ. Genome-wide analysis of self-reported risk-taking behaviour and cross-disorder genetic correlations in the UK Biobank cohort. Transl Psychiatry 2018; 8:39. [PMID: 29391395 PMCID: PMC5804026 DOI: 10.1038/s41398-017-0079-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 10/20/2017] [Accepted: 11/13/2017] [Indexed: 11/09/2022] Open
Abstract
Risk-taking behaviour is a key component of several psychiatric disorders and could influence lifestyle choices such as smoking, alcohol use, and diet. As a phenotype, risk-taking behaviour therefore fits within a Research Domain Criteria (RDoC) approach, whereby identifying genetic determinants of this trait has the potential to improve our understanding across different psychiatric disorders. Here we report a genome-wide association study in 116,255 UK Biobank participants who responded yes/no to the question "Would you consider yourself a risk taker?" Risk takers (compared with controls) were more likely to be men, smokers, and have a history of psychiatric disorder. Genetic loci associated with risk-taking behaviour were identified on chromosomes 3 (rs13084531) and 6 (rs9379971). The effects of both lead SNPs were comparable between men and women. The chromosome 3 locus highlights CADM2, previously implicated in cognitive and executive functions, but the chromosome 6 locus is challenging to interpret due to the complexity of the HLA region. Risk-taking behaviour shared significant genetic risk with schizophrenia, bipolar disorder, attention-deficit hyperactivity disorder, and post-traumatic stress disorder, as well as with smoking and total obesity. Despite being based on only a single question, this study furthers our understanding of the biology of risk-taking behaviour, a trait that has a major impact on a range of common physical and mental health disorders.
Collapse
Affiliation(s)
- Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Sarah Hartz
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Amy Horton
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO, USA
- Transmontane Analytics, Tuscon, AZ, USA
| | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Nicholas Graham
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Amy Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Daniel Mackay
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Laura M Pidgeon
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Jonathan Cavanagh
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Michael O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | | | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| |
Collapse
|
252
|
Johnsson M, Henriksen R, Höglund A, Fogelholm J, Jensen P, Wright D. Genetical genomics of growth in a chicken model. BMC Genomics 2018; 19:72. [PMID: 29361907 PMCID: PMC5782384 DOI: 10.1186/s12864-018-4441-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/08/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The genetics underlying body mass and growth are key to understanding a wide range of topics in biology, both evolutionary and developmental. Body mass and growth traits are affected by many genetic variants of small effect. This complicates genetic mapping of growth and body mass. Experimental intercrosses between individuals from divergent populations allows us to map naturally occurring genetic variants for selected traits, such as body mass by linkage mapping. By simultaneously measuring traits and intermediary molecular phenotypes, such as gene expression, one can use integrative genomics to search for potential causative genes. RESULTS In this study, we use linkage mapping approach to map growth traits (N = 471) and liver gene expression (N = 130) in an advanced intercross of wild Red Junglefowl and domestic White Leghorn layer chickens. We find 16 loci for growth traits, and 1463 loci for liver gene expression, as measured by microarrays. Of these, the genes TRAK1, OSBPL8, YEATS4, CEP55, and PIP4K2B are identified as strong candidates for growth loci in the chicken. We also show a high degree of sex-specific gene-regulation, with almost every gene expression locus exhibiting sex-interactions. Finally, several trans-regulatory hotspots were found, one of which coincides with a major growth locus. CONCLUSIONS These findings not only serve to identify several strong candidates affecting growth, but also show how sex-specificity and local gene-regulation affect growth regulation in the chicken.
Collapse
Affiliation(s)
- Martin Johnsson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, UK.,Department of Animal Breeding and Genetics, The Swedish University of Agricultural Sciences, Box 7023, 750 07, Uppsala, Sweden.,AVIAN Behavioural Genomics and Physiology Group, IFM Biology, Linköping University, 581 83, Linköping, Sweden
| | - Rie Henriksen
- AVIAN Behavioural Genomics and Physiology Group, IFM Biology, Linköping University, 581 83, Linköping, Sweden
| | - Andrey Höglund
- AVIAN Behavioural Genomics and Physiology Group, IFM Biology, Linköping University, 581 83, Linköping, Sweden
| | - Jesper Fogelholm
- AVIAN Behavioural Genomics and Physiology Group, IFM Biology, Linköping University, 581 83, Linköping, Sweden
| | - Per Jensen
- AVIAN Behavioural Genomics and Physiology Group, IFM Biology, Linköping University, 581 83, Linköping, Sweden
| | - Dominic Wright
- AVIAN Behavioural Genomics and Physiology Group, IFM Biology, Linköping University, 581 83, Linköping, Sweden.
| |
Collapse
|
253
|
Re-analysis of public genetic data reveals a rare X-chromosomal variant associated with type 2 diabetes. Nat Commun 2018; 9:321. [PMID: 29358691 PMCID: PMC5778074 DOI: 10.1038/s41467-017-02380-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 11/24/2017] [Indexed: 12/20/2022] Open
Abstract
The reanalysis of existing GWAS data represents a powerful and cost-effective opportunity to gain insights into the genetics of complex diseases. By reanalyzing publicly available type 2 diabetes (T2D) genome-wide association studies (GWAS) data for 70,127 subjects, we identify seven novel associated regions, five driven by common variants (LYPLAL1, NEUROG3, CAMKK2, ABO, and GIP genes), one by a low-frequency (EHMT2), and one driven by a rare variant in chromosome Xq23, rs146662075, associated with a twofold increased risk for T2D in males. rs146662075 is located within an active enhancer associated with the expression of Angiotensin II Receptor type 2 gene (AGTR2), a modulator of insulin sensitivity, and exhibits allelic specific activity in muscle cells. Beyond providing insights into the genetics and pathophysiology of T2D, these results also underscore the value of reanalyzing publicly available data using novel genetic resources and analytical approaches. Genome-wide association studies have uncovered several loci associated with diabetes risk. Here, the authors reanalyse public type 2 diabetes GWAS data to fine map 50 known loci and identify seven new ones, including one near ATGR2 on the X-chromosome that doubles the risk of diabetes in men.
Collapse
|
254
|
Gao C, Langefeld CD, Ziegler JT, Taylor KD, Norris JM, Chen YDI, Hellwege JN, Guo X, Allison MA, Speliotes EK, Rotter JI, Bowden DW, Wagenknecht LE, Palmer ND. Genome-Wide Study of Subcutaneous and Visceral Adipose Tissue Reveals Novel Sex-Specific Adiposity Loci in Mexican Americans. Obesity (Silver Spring) 2018; 26:202-212. [PMID: 29178545 PMCID: PMC5740005 DOI: 10.1002/oby.22074] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 01/02/2023]
Abstract
OBJECTIVE This study aimed to explore the genetic mechanisms of regional fat deposition, which is a strong risk factor for metabolic diseases beyond total adiposity. METHODS A genome-wide association study of 7,757,139 single-nucleotide polymorphisms (SNPs) in 983 Mexican Americans (nmale = 403; nfemale = 580) from the Insulin Resistance Atherosclerosis Family Study was performed. Association analyses were performed with and without sex stratification for subcutaneous adipose tissue, visceral adipose tissue (VAT), and visceral-subcutaneous ratio (VSR) obtained from computed tomography. RESULTS The strongest signal identified was SNP rs2185405 (minor allele frequencies [MAF] = 40%; PVAT = 1.98 × 10-8 ) with VAT. It is an intronic variant of the GLIS family zinc finger 3 gene (GLIS3). In addition, SNP rs12657394 (MAF = 19%) was associated with VAT in males (Pmale = 2.39×10-8 ; Pfemale = 2.5 × 10-3 ). It is located intronically in the serum response factor binding protein 1 gene (SRFBP1). On average, male carriers of the variant had 24.6 cm2 increased VAT compared with noncarriers. Subsequently, genome-wide SNP-sex interaction analysis was performed. SNP rs10913233 (MAF = 14%; Pint = 3.07 × 10-8 ) in PAPPA2 and rs10923724 (MAF = 38%; Pint = 2.89 × 10-8 ) upstream of TBX15 were strongly associated with the interaction effect for VSR. CONCLUSIONS Six loci were identified with genome-wide significant associations with fat deposition and interactive effects. These results provided genetic evidence for a differential basis of fat deposition between genders.
Collapse
Affiliation(s)
- Chuan Gao
- Molecular Genetics and Genomics Program; Wake Forest School of Medicine, Winston-Salem, NC
- Center for Genomics and Personalized Medicine Research; Wake Forest School of Medicine, Winston-Salem, NC
- Center for Public Health Genomics; Wake Forest School of Medicine, Winston-Salem, NC
| | - Carl D. Langefeld
- Center for Public Health Genomics; Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biostatistical Sciences; Wake Forest School of Medicine, Winston-Salem, NC
| | - Julie T. Ziegler
- Center for Public Health Genomics; Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biostatistical Sciences; Wake Forest School of Medicine, Winston-Salem, NC
| | - Kent D. Taylor
- Institute for Translational Genomics and Population Sciences; Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health; University of Colorado, Aurora, CO
| | - Yii-Der I. Chen
- Institute for Translational Genomics and Population Sciences; Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Jacklyn N. Hellwege
- Center for Genomics and Personalized Medicine Research; Wake Forest School of Medicine, Winston-Salem, NC
- Center for Diabetes Research; Wake Forest School of Medicine, Winston-Salem, NC
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences; Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Matthew A. Allison
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla CA
| | - Elizabeth K. Speliotes
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics; University of Michigan, Ann Arbor, MI
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences; Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
- Department of Pediatrics; Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Donald W. Bowden
- Center for Genomics and Personalized Medicine Research; Wake Forest School of Medicine, Winston-Salem, NC
- Center for Diabetes Research; Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biochemistry; Wake Forest School of Medicine, Winston-Salem, NC
| | - Lynne E. Wagenknecht
- Division of Public Health Sciences; Wake Forest School of Medicine, Winston-Salem, NC
| | - Nicholette D. Palmer
- Center for Genomics and Personalized Medicine Research; Wake Forest School of Medicine, Winston-Salem, NC
- Center for Public Health Genomics; Wake Forest School of Medicine, Winston-Salem, NC
- Center for Diabetes Research; Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biochemistry; Wake Forest School of Medicine, Winston-Salem, NC
- Correspondence to Nicholette D. Palmer, PhD, Department of Biochemistry, 1 Medical Center Blvd, Winston-Salem, NC 27040, Phone: 336-713-7534,
| |
Collapse
|
255
|
Johnson SC. Nutrient Sensing, Signaling and Ageing: The Role of IGF-1 and mTOR in Ageing and Age-Related Disease. Subcell Biochem 2018; 90:49-97. [PMID: 30779006 DOI: 10.1007/978-981-13-2835-0_3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nutrient signaling through insulin/IGF-1 was the first pathway demonstrated to regulate ageing and age-related disease in model organisms. Pharmacological or dietary interventions targeting nutrient signaling pathways have been shown to robustly attenuate ageing in many organisms. Caloric restriction, the most widely studied longevity promoting intervention, works through multiple nutrient signaling pathways, while inhibition of mTOR through treatment with rapamycin reproducibly delays ageing and disease through specific inhibition of the mTOR complexes. Although the benefits of reduced insulin/IGF-1 in lifespan and health are well documented in model organisms, defining the precise role of the IGF-1 in human ageing and age-related disease has proven more difficult. Association studies provide some insight but also reveal paradoxes. Low serum IGF-1 predicts longevity, but IGF-1 decreases with age and IGF-1 therapy benefits some of age-related pathologies. Circulating IGF-1 has been associated both positively and negatively with risk of age-related diseases in humans, and in some cases both activation and inhibition of IGF-1 signaling have provided benefit in animal models of the same diseases. Interventions designed modulate the nutrient sensing signaling pathways positively or negatively are already available for clinical use, highlighting the need for a clear understanding of the role of nutrient signaling in ageing and age-related disease. This chapter examines data from model organisms and human genetic association studies, with a special emphasis on IGF-1 and mTOR, and discusses potential models for resolving the paradoxes surrounding IGF-1 data.
Collapse
Affiliation(s)
- Simon C Johnson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.
| |
Collapse
|
256
|
Raghow R. Gut-brain crosstalk regulates craving for fatty food. World J Diabetes 2017; 8:484-488. [PMID: 29290921 PMCID: PMC5740093 DOI: 10.4239/wjd.v8.i12.484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/20/2017] [Accepted: 09/04/2017] [Indexed: 02/05/2023] Open
Abstract
Patients undergoing Roux-en-Y gastric bypass (RYGB) surgery elicit striking loss of body weight. Anatomical re-structuring of the gastrointestinal (GI) tract, leading to reduced caloric intake and changes in food preference, are thought to be the primary drivers of weight loss in bariatric surgery patients. However, the mechanisms by which RYGB surgery causes a reduced preference for fatty foods remain elusive. In a recent report, Hankir et al described how RYGB surgery modulated lipid nutrient signals in the intestine of rats to blunt their craving for fatty food. The authors reported that RYGB surgery restored an endogenous fat-satiety signaling pathway, mediated via oleoylethanolamide (OEA), that was greatly blunted in obese animals. In RYGB rats, high fat diet (HFD) led to increased production of OEA that activated the intestinal peroxisome proliferation activator receptors-α (PPARα). In RYGB rats, activation of PPARα by OEA was accompanied by enhanced dopamine neurotransmission in the dorsal striatum and reduced preference for HFD. The authors showed that OEA-mediated signals to the midbrain were transmitted via the vagus nerve. Interfering with either the production of OEA in enterocytes, or blocking of vagal and striatal D1 receptors signals eliminated the decreased craving for fat in RYGB rats. These studies demonstrated that bariatric surgery led to alterations in the reward circuitry of the brain in RYGB rats and reduced their preference for HFD.
Collapse
Affiliation(s)
- Rajendra Raghow
- Department of Veterans Affairs Medical Center, Memphis, TN 38104, United States
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| |
Collapse
|
257
|
Abadi A, Alyass A, Robiou du Pont S, Bolker B, Singh P, Mohan V, Diaz R, Engert JC, Yusuf S, Gerstein HC, Anand SS, Meyre D. Penetrance of Polygenic Obesity Susceptibility Loci across the Body Mass Index Distribution. Am J Hum Genet 2017; 101:925-938. [PMID: 29220676 PMCID: PMC5812888 DOI: 10.1016/j.ajhg.2017.10.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022] Open
Abstract
A growing number of single-nucleotide polymorphisms (SNPs) have been associated with body mass index (BMI) and obesity, but whether the effects of these obesity-susceptibility loci are uniform across the BMI distribution remains unclear. We studied the effects of 37 BMI-associated SNPs in 75,230 adults of European ancestry across BMI percentiles by using conditional quantile regression (CQR) and meta-regression (MR) models. The effects of nine SNPs (24%)-rs1421085 (FTO; p = 8.69 × 10-15), rs6235 (PCSK1; p = 7.11 × 10-6), rs7903146 (TCF7L2; p = 9.60 × 10-6), rs11873305 (MC4R; p = 5.08 × 10-5), rs12617233 (FANCL; p = 5.30 × 10-5), rs11672660 (GIPR; p = 1.64 × 10-4), rs997295 (MAP2K5; p = 3.25 × 10-4), rs6499653 (FTO; p = 6.23 × 10-4), and rs3824755 (NT5C2; p = 7.90 × 10-4)-increased significantly across the sample BMI distribution. We showed that such increases stemmed from unadjusted gene interactions that enhanced the effects of SNPs in persons with a high BMI. When 125 height-associated SNPs were analyzed for comparison, only one (<1%), rs6219 (IGF1, p = 1.80 × 10-4), showed effects that varied significantly across height percentiles. Cumulative gene scores of these SNPs (GS-BMI and GS-height) showed that only GS-BMI had effects that increased significantly across the sample distribution (BMI: p = 7.03 × 10-37; height: p = 0.499). Overall, these findings underscore the importance of gene-gene and gene-environment interactions in shaping the genetic architecture of BMI and advance a method for detecting such interactions by using only the sample outcome distribution.
Collapse
Affiliation(s)
- Arkan Abadi
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Akram Alyass
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sebastien Robiou du Pont
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Ben Bolker
- Department of Mathematics and Statistics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Pardeep Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation, Gopalapuram, Chennai 600086, India
| | - Rafael Diaz
- Estudios Clínicos Latino America, Paraguay 160, S2000CVD Rosario, Santa Fe, Argentina
| | | | - Salim Yusuf
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton General Hospital, Hamilton, ON L8S 4L8, Canada; Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Hertzel C Gerstein
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton General Hospital, Hamilton, ON L8S 4L8, Canada; Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sonia S Anand
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton General Hospital, Hamilton, ON L8S 4L8, Canada; Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
258
|
van Waateringe RP, Muller Kobold AC, van Vliet-Ostaptchouk JV, van der Klauw MM, Koerts J, Anton G, Peters A, Trischler G, Kvaløy K, Naess M, Videm V, Hveem K, Waldenberger M, Koenig W, Wolffenbuttel BH. Influence of Storage and Inter- and Intra-Assay Variability on the Measurement of Inflammatory Biomarkers in Population-Based Biobanking. Biopreserv Biobank 2017; 15:512-518. [DOI: 10.1089/bio.2017.0001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Anneke C. Muller Kobold
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Jan Koerts
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gabriele Anton
- Research Unit of Molecular Epidemiology and Institute of Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Annette Peters
- Research Unit of Molecular Epidemiology and Institute of Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Gerlinde Trischler
- Department of Internal Medicine II–Cardiology, University of Ulm Medical Centre, Ulm, Germany
| | - Kirsti Kvaløy
- Department of Public Health and General Practice, Faculty of Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Naess
- Department of Public Health and General Practice, Faculty of Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Vibeke Videm
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Kristian Hveem
- Department of Public Health and General Practice, Faculty of Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology and Institute of Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II–Cardiology, University of Ulm Medical Centre, Ulm, Germany
| | | |
Collapse
|
259
|
Martínez-Barquero V, Marco GD, Martínez-Hervas S, Adam-Felici V, Pérez-Soriano C, Gonzalez-Albert V, Rojo G, Ascaso JF, Real JT, Garcia-Garcia AB, Martín-Escudero JC, Cortes R, Chaves FJ. Are IL18RAP gene polymorphisms associated with body mass regulation? A cross-sectional study. BMJ Open 2017; 7:e017875. [PMID: 29146643 PMCID: PMC5695454 DOI: 10.1136/bmjopen-2017-017875] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVES To investigate the association between IL18RAP and body mass index (BMI) and obesity and to verify the effect of a polymorphism in the microRNA136 (MIR136) IL18RAP binding region. DESIGN We analysed samples from two Spanish cross-sectional studies, VALCAR (Spanish Mediterranean coast) and Hortega (Spanish centre). These studies aimed at analysing cardiovascular risk and development of cardiovascular disease in the general population. Both populations correspond to regions with different characteristics. SETTING Five IL18RAP single nucleotide polymorphisms were selected using the SYSNPs web tool and analysed by oligonucleotide ligation assay (SNPlex). For the MIR136 functional study, cells were transfected with plasmids containing different rs7559479 polymorphism alleles and analysed by luciferase reporter assays. PARTICIPANTS 1970 individuals (Caucasian, both genders): VALCAR (468) and Hortega (1502). RESULTS rs2293225, rs2272127 and rs7559479 showed the following associations: rs7559479 G allele correlated with a higher obesity risk (P=0.01; OR=1.82; 95% CI 1.15 to 2.87 for the VALCAR group; P=0.033; OR=1.35; 95% CI 1.03 to 1.79 for the Hortega population) and higher body mass index (BMI) values (P=0.0045; P=0.1 for VALCAR and Hortega, respectively); a significant association with obesity (P=0.0024, OR=1.44, 95% CI 1.14 to 1.82) and increased BMI values (P=0.008) was found when considering both populations together. rs2293225 T allele was associated with lower obesity risk (P=0.036; OR=0.60; 95% CI 0.35 to 0.96) and lower BMI values (P=0.0038; OR=1.41) while the rs2272127 G allele was associated with lower obesity risk (P=0.028; OR=0.66; 95% CI 0.44 to 0.97) only in the VALCAR population. A reporter assay showed that the presence of the A allele in rs7559479 was associated with increased MIR136 binding to IL18RAP. CONCLUSIONS Our results suggest that polymorphisms in IL18RAP influence susceptibility to obesity. We demonstrated that the A allele in rs7559479 increases MIR136 binding, which regulates IL-18 system activity.
Collapse
Affiliation(s)
- Vanesa Martínez-Barquero
- Department of Medicine, University of Valencia, Valencia, Spain
- Department of Genomic and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Griselda de Marco
- Department of Genomic 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), Madrid, Spain
- Department of Endocrinology and Nutrition Service, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Victoria Adam-Felici
- Department of Genomic and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Cristina Pérez-Soriano
- Department of Genomic and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Verónica Gonzalez-Albert
- Department of Genomic and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Gemma Rojo
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
- Department of Endocrinology and Nutrition Service, Hospital Regional Universitario, Instituto de Biomedicina de Málaga (IBIMA), Malaga, Spain
| | - Juan Francisco Ascaso
- Department of Medicine, University of Valencia, Valencia, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
- Department of Endocrinology and Nutrition Service, 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), Madrid, Spain
- Department of Endocrinology and Nutrition Service, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Ana Barbara Garcia-Garcia
- Department of Genomic and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | | | - Raquel Cortes
- Department of Genomic and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
| | - Felipe Javier Chaves
- Department of Genomic and Genetic Diagnosis Unit, Hospital Clínico Research Foundation (INCLIVA), Valencia, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| |
Collapse
|
260
|
Lo MT, Wang Y, Kauppi K, Sanyal N, Fan CC, Smeland OB, Schork A, Holland D, Hinds DA, Tung JY, Andreassen OA, Dale AM, Chen CH. Modeling prior information of common genetic variants improves gene discovery for neuroticism. Hum Mol Genet 2017; 26:4530-4539. [PMID: 28973307 PMCID: PMC5886256 DOI: 10.1093/hmg/ddx340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/10/2017] [Accepted: 08/25/2017] [Indexed: 02/01/2023] Open
Abstract
Neuroticism reflects emotional instability, and is related to various mental and physical health issues. However, the majority of genetic variants associated with neuroticism remain unclear. Inconsistent genetic variants identified by different genome-wide association studies (GWAS) may be attributable to low statistical power. We proposed a novel framework to improve the power for gene discovery by incorporating prior information of single nucleotide polymorphisms (SNPs) and combining two relevant existing tools, relative enrichment score (RES) and conditional false discovery rate (FDR). Here, SNP's conditional FDR was estimated given its RES based on SNP prior information including linkage disequilibrium (LD)-weighted genic annotation scores, total LD scores and heterozygosity. A known significant locus in chromosome 8p was excluded before estimating FDR due to long-range LD structure. Only one significant LD-independent SNP was detected by analyses of unconditional FDR and traditional GWAS in the discovery sample (N = 59 225), and notably four additional SNPs by conditional FDR. Three of the five SNPs, all identified by conditional FDR, were replicated (P < 0.05) in an independent sample (N = 170 911). These three SNPs are located in intronic regions of CADM2, LINGO2 and EP300 which have been reported to be associated with autism, Parkinson's disease and schizophrenia, respectively. Our approach using a combination of RES and conditional FDR improved power of traditional GWAS for gene discovery providing a useful framework for the analysis of GWAS summary statistics by utilizing SNP prior information, and helping to elucidate the links between neuroticism and complex diseases from a genetic perspective.
Collapse
Affiliation(s)
- Min-Tzu Lo
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA 92037, USA
| | - Yunpeng Wang
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo 0407, Norway
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Karolina Kauppi
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA 92037, USA
- Department of Radiation Sciences, Umea University, Umea 90187, Sweden
| | - Nilotpal Sanyal
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA 92037, USA
| | - Chun-Chieh Fan
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA 92037, USA
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA 92037, USA
| | - Olav B Smeland
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo 0407, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo 0407, Norway
| | - Andrew Schork
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA 92037, USA
- Institute of Biological Psychiatry, Medical Health Center, Sct. Hans, Roskilde, 4000, Denmark
| | - Dominic Holland
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92037, USA
| | | | | | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo 0407, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo 0407, Norway
| | - Anders M Dale
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA 92037, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92037, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92037, USA
| | - Chi-Hua Chen
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
261
|
Mao L, Fang Y, Campbell M, Southerland WM. Population differentiation in allele frequencies of obesity-associated SNPs. BMC Genomics 2017; 18:861. [PMID: 29126384 PMCID: PMC5681842 DOI: 10.1186/s12864-017-4262-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/02/2017] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Obesity is emerging as a global health problem, with more than one-third of the world's adult population being overweight or obese. In this study, we investigated worldwide population differentiation in allele frequencies of obesity-associated SNPs (single nucleotide polymorphisms). RESULTS We collected a total of 225 obesity-associated SNPs from a public database. Their population-level allele frequencies were derived based on the genotype data from 1000 Genomes Project (phase 3). We used hypergeometric model to assess whether the effect allele at a given SNP is significantly enriched or depleted in each of the 26 populations surveyed in the 1000 Genomes Project with respect to the overall pooled population. Our results indicate that 195 out of 225 SNPs (86.7%) possess effect alleles significantly enriched or depleted in at least one of the 26 populations. Populations within the same continental group exhibit similar allele enrichment/depletion patterns whereas inter-continental populations show distinct patterns. Among the 225 SNPs, 15 SNPs cluster in the first intron region of the FTO gene, which is a major gene associated with body-mass index (BMI) and fat mass. African populations exhibit much smaller blocks of LD (linkage disequilibrium) among these15 SNPs while European and Asian populations have larger blocks. To estimate the cumulative effect of all variants associated with obesity, we developed the personal composite genetic risk score for obesity. Our results indicate that the East Asian populations have the lowest averages of the composite risk scores, whereas three European populations have the highest averages. In addition, the population-level average of composite genetic risk scores is significantly correlated (R2 = 0.35, P = 0.0060) with obesity prevalence. CONCLUSIONS We have detected substantial population differentiation in allele frequencies of obesity-associated SNPs. The results will help elucidate the genetic basis which may contribute to population disparities in obesity prevalence.
Collapse
Affiliation(s)
- Linyong Mao
- Department of Biochemistry and Molecular Biology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059 USA
| | - Yayin Fang
- Department of Biochemistry and Molecular Biology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059 USA
| | - Michael Campbell
- Department of Biology, Howard University, 415 College Street NW, Washington, 20059 DC USA
| | - William M. Southerland
- Department of Biochemistry and Molecular Biology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059 USA
| |
Collapse
|
262
|
Calderon D, Bhaskar A, Knowles DA, Golan D, Raj T, Fu AQ, Pritchard JK. Inferring Relevant Cell Types for Complex Traits by Using Single-Cell Gene Expression. Am J Hum Genet 2017; 101:686-699. [PMID: 29106824 PMCID: PMC5673624 DOI: 10.1016/j.ajhg.2017.09.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/13/2017] [Indexed: 01/09/2023] Open
Abstract
Previous studies have prioritized trait-relevant cell types by looking for an enrichment of genome-wide association study (GWAS) signal within functional regions. However, these studies are limited in cell resolution by the lack of functional annotations from difficult-to-characterize or rare cell populations. Measurement of single-cell gene expression has become a popular method for characterizing novel cell types, and yet limited work has linked single-cell RNA sequencing (RNA-seq) to phenotypes of interest. To address this deficiency, we present RolyPoly, a regression-based polygenic model that can prioritize trait-relevant cell types and genes from GWAS summary statistics and gene expression data. RolyPoly is designed to use expression data from either bulk tissue or single-cell RNA-seq. In this study, we demonstrated RolyPoly's accuracy through simulation and validated previously known tissue-trait associations. We discovered a significant association between microglia and late-onset Alzheimer disease and an association between schizophrenia and oligodendrocytes and replicating fetal cortical cells. Additionally, RolyPoly computes a trait-relevance score for each gene to reflect the importance of expression specific to a cell type. We found that differentially expressed genes in the prefrontal cortex of individuals with Alzheimer disease were significantly enriched with genes ranked highly by RolyPoly gene scores. Overall, our method represents a powerful framework for understanding the effect of common variants on cell types contributing to complex traits.
Collapse
Affiliation(s)
- Diego Calderon
- Program in Biomedical Informatics, Stanford University, Stanford, CA 94305, USA.
| | - Anand Bhaskar
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David A Knowles
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - David Golan
- Faculty of Industrial Engineering & Management, Technion, Haifa 3200003, Israel
| | - Towfique Raj
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Audrey Q Fu
- Department of Statistical Science, University of Idaho, Moscow, ID 83844, USA
| | - Jonathan K Pritchard
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
263
|
Gene-nutrient interactions and susceptibility to human obesity. GENES AND NUTRITION 2017; 12:29. [PMID: 29093760 PMCID: PMC5663124 DOI: 10.1186/s12263-017-0581-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/04/2017] [Indexed: 12/28/2022]
Abstract
A large number of genome-wide association studies, transferability studies, and candidate gene studies performed in diverse populations around the world have identified gene variants that are associated with common human obesity. The mounting evidence suggests that these obesity gene variants interact with multiple environmental factors and increase susceptibility to this complex metabolic disease. The objective of this review article is to provide concise and updated information on energy balance, heritability of body weight, origins of gene variants, and gene-nutrient interactions in relation to human obesity. It is proposed that knowledge of these related topics will provide valuable insight for future preventative lifestyle intervention using targeted nutritional and medicinal therapies.
Collapse
|
264
|
Multiphenotype association study of patients randomized to initiate antiretroviral regimens in AIDS Clinical Trials Group protocol A5202. Pharmacogenet Genomics 2017; 27:101-111. [PMID: 28099408 PMCID: PMC5285297 DOI: 10.1097/fpc.0000000000000263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Supplemental Digital Content is available in the text. Background High-throughput approaches are increasingly being used to identify genetic associations across multiple phenotypes simultaneously. Here, we describe a pilot analysis that considered multiple on-treatment laboratory phenotypes from antiretroviral therapy-naive patients who were randomized to initiate antiretroviral regimens in a prospective clinical trial, AIDS Clinical Trials Group protocol A5202. Participants and methods From among 5 9545 294 polymorphisms imputed genome-wide, we analyzed 2544, including 2124 annotated in the PharmGKB, and 420 previously associated with traits in the GWAS Catalog. We derived 774 phenotypes on the basis of context from six variables: plasma atazanavir (ATV) pharmacokinetics, plasma efavirenz (EFV) pharmacokinetics, change in the CD4+ T-cell count, HIV-1 RNA suppression, fasting low-density lipoprotein-cholesterol, and fasting triglycerides. Permutation testing assessed the likelihood of associations being by chance alone. Pleiotropy was assessed for polymorphisms with the lowest P-values. Results This analysis included 1181 patients. At P less than 1.5×10−4, most associations were not by chance alone. Polymorphisms with the lowest P-values for EFV pharmacokinetics (CYPB26 rs3745274), low-density lipoprotein -cholesterol (APOE rs7412), and triglyceride (APOA5 rs651821) phenotypes had been associated previously with those traits in previous studies. The association between triglycerides and rs651821 was present with ATV-containing regimens, but not with EFV-containing regimens. Polymorphisms with the lowest P-values for ATV pharmacokinetics, CD4 T-cell count, and HIV-1 RNA phenotypes had not been reported previously to be associated with that trait. Conclusion Using data from a prospective HIV clinical trial, we identified expected genetic associations, potentially novel associations, and at least one context-dependent association. This study supports high-throughput strategies that simultaneously explore multiple phenotypes from clinical trials’ datasets for genetic associations.
Collapse
|
265
|
Paré G, Mao S, Deng WQ. A machine-learning heuristic to improve gene score prediction of polygenic traits. Sci Rep 2017; 7:12665. [PMID: 28979001 PMCID: PMC5627249 DOI: 10.1038/s41598-017-13056-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 09/18/2017] [Indexed: 01/25/2023] Open
Abstract
Machine-learning techniques have helped solve a broad range of prediction problems, yet are not widely used to build polygenic risk scores for the prediction of complex traits. We propose a novel heuristic based on machine-learning techniques (GraBLD) to boost the predictive performance of polygenic risk scores. Gradient boosted regression trees were first used to optimize the weights of SNPs included in the score, followed by a novel regional adjustment for linkage disequilibrium. A calibration set with sample size of ~200 individuals was sufficient for optimal performance. GraBLD yielded prediction R2 of 0.239 and 0.082 using GIANT summary association statistics for height and BMI in the UK Biobank study (N = 130 K; 1.98 M SNPs), explaining 46.9% and 32.7% of the overall polygenic variance, respectively. For diabetes status, the area under the receiver operating characteristic curve was 0.602 in the UK Biobank study using summary-level association statistics from the DIAGRAM consortium. GraBLD outperformed other polygenic score heuristics for the prediction of height (p < 2.2 × 10−16) and BMI (p < 1.57 × 10−4), and was equivalent to LDpred for diabetes. Results were independently validated in the Health and Retirement Study (N = 8,292; 688,398 SNPs). Our report demonstrates the use of machine-learning techniques, coupled with summary-level data from large genome-wide meta-analyses to improve the prediction of polygenic traits.
Collapse
Affiliation(s)
- Guillaume Paré
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Canada. .,Population Genomics Program, Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Canada. .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.
| | - Shihong Mao
- 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
|
266
|
Iacomino G, Siani A. Role of microRNAs in obesity and obesity-related diseases. GENES AND NUTRITION 2017; 12:23. [PMID: 28974990 PMCID: PMC5613467 DOI: 10.1186/s12263-017-0577-z] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/12/2017] [Indexed: 12/15/2022]
Abstract
In recent years, the link between regulatory microRNAs (miRNAs) and diseases has been the object of intensive research. miRNAs have emerged as key mediators of metabolic processes, playing crucial roles in maintaining/altering physiological processes, including energy balance and metabolic homeostasis. Altered miRNAs expression has been reported in association with obesity, both in animal and human studies. Dysregulation of miRNAs may affect the status and functions of different tissues and organs, including the adipose tissue, pancreas, liver, and muscle, possibly contributing to metabolic abnormalities associated with obesity and obesity-related diseases. More recently, the discovery of circulating miRNAs easily detectable in plasma and other body fluids has emphasized their potential as both endocrine signaling molecules and disease indicators. In this review, the status of current research on the role of miRNAs in obesity and related metabolic abnormalities is summarized and discussed.
Collapse
Affiliation(s)
- Giuseppe Iacomino
- Institute of Food Sciences, CNR, Via Roma, 64, 83100 Avellino, Italy
| | - Alfonso Siani
- Institute of Food Sciences, CNR, Via Roma, 64, 83100 Avellino, Italy
| |
Collapse
|
267
|
Klarin D, Zhu QM, Emdin CA, Chaffin M, Horner S, McMillan BJ, Leed A, Weale ME, Spencer CCA, Aguet F, Segrè AV, Ardlie KG, Khera AV, Kaushik VK, Natarajan P, Consortium CARDI, Kathiresan S. Genetic analysis in UK Biobank links insulin resistance and transendothelial migration pathways to coronary artery disease. Nat Genet 2017; 49:1392-1397. [PMID: 28714974 PMCID: PMC5577383 DOI: 10.1038/ng.3914] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/15/2017] [Indexed: 12/17/2022]
Abstract
UK Biobank is among the world's largest repositories for phenotypic and genotypic information in individuals of European ancestry. We performed a genome-wide association study in UK Biobank testing ∼9 million DNA sequence variants for association with coronary artery disease (4,831 cases and 115,455 controls) and carried out meta-analysis with previously published results. We identified 15 new loci, bringing the total number of loci associated with coronary artery disease to 95 at the time of analysis. Phenome-wide association scanning showed that CCDC92 likely affects coronary artery disease through insulin resistance pathways, whereas experimental analysis suggests that ARHGEF26 influences the transendothelial migration of leukocytes.
Collapse
Affiliation(s)
- Derek Klarin
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston MA, USA
| | - Qiuyu Martin Zhu
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
| | - Connor A. Emdin
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
| | - Mark Chaffin
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
| | - Steven Horner
- Center for the Development of Therapeutics, Broad Institute, Cambridge MA, USA
| | - Brian J. McMillan
- Center for the Development of Therapeutics, Broad Institute, Cambridge MA, USA
| | - Alison Leed
- Center for the Development of Therapeutics, Broad Institute, Cambridge MA, USA
| | | | | | | | | | - Kristin G. Ardlie
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
| | - Amit V. Khera
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
| | - Virendar K. Kaushik
- Center for the Development of Therapeutics, Broad Institute, Cambridge MA, USA
| | - Pradeep Natarajan
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
| | | | - Sekar Kathiresan
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge MA, USA
| |
Collapse
|
268
|
Inflammation and bone mineral density: A Mendelian randomization study. Sci Rep 2017; 7:8666. [PMID: 28819125 PMCID: PMC5561220 DOI: 10.1038/s41598-017-09080-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/21/2017] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis is a common age-related disorder leading to an increase in osteoporotic fractures and resulting in significant suffering and disability. Inflammation may contribute to osteoporosis, as it does to many other chronic diseases. We examined whether inflammation is etiologically relevant to osteoporosis, assessed from bone mineral density (BMD), as a new potential target of intervention, or whether it is a symptom/biomarker of osteoporosis. We obtained genetic predictors of inflammatory markers from genome-wide association studies and applied them to a large genome wide association study of BMD. Using two-sample Mendelian randomization, we obtained unconfounded estimates of the effect of high-sensitivity C-reactive protein (hsCRP) on BMD at the forearm, femoral neck, and lumbar spine. After removing potentially pleiotropic single nucleotide polymorphisms (SNPs) possibly acting via obesity-related traits, hsCRP, based on 16 SNPs from genes including CRP, was not associated with BMD. A causal relation of hsCRP with lower BMD was not evident in this study.
Collapse
|
269
|
Butler JM, Hall N, Narendran N, Yang YC, Paraoan L. Identification of candidate protective variants for common diseases and evaluation of their protective potential. BMC Genomics 2017; 18:575. [PMID: 28774272 PMCID: PMC5543444 DOI: 10.1186/s12864-017-3964-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 07/27/2017] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Human polymorphisms with derived alleles that are protective against disease may provide powerful translational opportunities. Here we report a method to identify such candidate polymorphisms and apply it to common non-synonymous SNPs (nsSNPs) associated with common diseases. Our study also sought to establish which of the identified protective nsSNPs show evidence of positive selection, taking this as indirect evidence that the protective variant has a beneficial effect on phenotype. Further, we performed an analysis to quantify the predicted effect of each protective variant on protein function/structure. RESULTS An initial analysis of eight SNPs previously identified as associated with age-related macular degeneration (AMD), revealed that two of them have a derived allele that is protective against developing the disease. One is in the complement component 2 gene (C2; E318D) and the other is in the complement factor B gene (CFB; R32Q). Then, combining genomewide ancestral allele information with known common disease-associated nsSNPs from the GWAS catalog, we found 32 additional SNPs which have a derived allele that is disease protective. Out of the total 34 identified candidate protective variants (CPVs), we found that 30 show stronger evidence of positive selection than the protective variant in lipoprotein lipase (LPL; S447X), which has already been translated into gene therapy. Furthermore, 11 of these CPVs have a higher probability of affecting protein structure than the lipoprotein lipase protective variant (LPL; S447X). CONCLUSIONS We identify 34 CPVs from the human genome. Diseases they confer protection against include, but are not limited to, type 2 diabetes, inflammatory bowel disease, age-related macular degeneration, multiple sclerosis and rheumatoid arthritis. We propose that those 30 CPVs with evidence of stronger positive selection than the LPL protective variant, may be considered as priority candidates for therapeutic approaches. The next step towards translation will require testing the hypotheses generated by our analyses, specifically whether the CPV arose from a gain-of-function or a loss-of-function mutation.
Collapse
Affiliation(s)
- Joe M Butler
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Neil Hall
- The Earlham Institute, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Niro Narendran
- Department of Ophthalmology, The Royal Wolverhampton NHS Trust, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Yit C Yang
- Department of Ophthalmology, The Royal Wolverhampton NHS Trust, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Luminita Paraoan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, 6 West Derby Street, Liverpool, L7 8TX, UK.
| |
Collapse
|
270
|
Coram MA, Fang H, Candille SI, Assimes TL, Tang H. Leveraging Multi-ethnic Evidence for Risk Assessment of Quantitative Traits in Minority Populations. Am J Hum Genet 2017; 101:218-226. [PMID: 28757202 DOI: 10.1016/j.ajhg.2017.06.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/29/2017] [Indexed: 11/20/2022] Open
Abstract
An essential component of precision medicine is the ability to predict an individual's risk of disease based on genetic and non-genetic factors. For complex traits and diseases, assessing the risk due to genetic factors is challenging because it requires knowledge of both the identity of variants that influence the trait and their corresponding allelic effects. Although the set of risk variants and their allelic effects may vary between populations, a large proportion of these variants were identified based on studies in populations of European descent. Heterogeneity in genetic architecture underlying complex traits and diseases, while broadly acknowledged, remains poorly characterized. Ignoring such heterogeneity likely reduces predictive accuracy for minority individuals. In this study, we propose an approach, called XP-BLUP, which ameliorates this ethnic disparity by combining trans-ethnic and ethnic-specific information. We build a polygenic model for complex traits that distinguishes candidate trait-relevant variants from the rest of the genome. The set of candidate variants are selected based on studies in any human population, yet the allelic effects are evaluated in a population-specific fashion. Simulation studies and real data analyses demonstrate that XP-BLUP adaptively utilizes trans-ethnic information and can substantially improve predictive accuracy in minority populations. At the same time, our study highlights the importance of the continued expansion of minority cohorts.
Collapse
Affiliation(s)
- Marc A Coram
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Huaying Fang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sophie I Candille
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Themistocles L Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hua Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
271
|
Pettersson M, Viljakainen H, Loid P, Mustila T, Pekkinen M, Armenio M, Andersson-Assarsson JC, Mäkitie O, Lindstrand A. Copy Number Variants Are Enriched in Individuals With Early-Onset Obesity and Highlight Novel Pathogenic Pathways. J Clin Endocrinol Metab 2017; 102:3029-3039. [PMID: 28605459 DOI: 10.1210/jc.2017-00565] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/07/2017] [Indexed: 01/22/2023]
Abstract
CONTEXT Only a few genetic causes for childhood obesity have been identified to date. Copy number variants (CNVs) are known to contribute to obesity, both syndromic (15q11.2 deletions, Prader-Willi syndrome) and nonsyndromic (16p11.2 deletions) obesity. OBJECTIVE To study the contribution of CNVs to early-onset obesity and evaluate the expression of candidate genes in subcutaneous adipose tissue. DESIGN AND SETTING A case-control study in a tertiary academic center. PARTICIPANTS CNV analysis was performed on 90 subjects with early-onset obesity and 67 normal-weight controls. Subcutaneous adipose tissue from body mass index-discordant siblings was used for the gene expression analyses. MAIN OUTCOME MEASURES We used custom high-density array comparative genomic hybridization with exon resolution in 1989 genes, including all known obesity loci. The expression of candidate genes was assessed using microarray analysis of messenger RNA from subcutaneous adipose tissue. RESULTS We identified rare CNVs in 17 subjects (19%) with obesity and 2 controls (3%). In three cases (3%), the identified variant involved a known syndromic lesion (22q11.21 duplication, 1q21.1 deletion, and 16p11.2 deletion, respectively), although the others were not known. Seven CNVs in 10 families were inherited and segregated with obesity. Expression analysis of 37 candidate genes showed discordant expression for 10 genes (PCM1, EFEMP1, MAMLD1, ACP6, BAZ2B, SORBS1, KLF15, MACROD2, ATR, and MBD5). CONCLUSIONS Rare CNVs contribute possibly pathogenic alleles to a substantial fraction of children with early-onset obesity. The involved genes might provide insights into pathogenic mechanisms and involved cellular pathways. These findings highlight the importance of CNV screening in children with early-onset obesity.
Collapse
MESH Headings
- Abnormalities, Multiple/genetics
- Acid Phosphatase/genetics
- Adolescent
- Adult
- Ataxia Telangiectasia Mutated Proteins/genetics
- Autistic Disorder/genetics
- Autoantigens/genetics
- Case-Control Studies
- Cell Cycle Proteins/genetics
- Child
- Child, Preschool
- Chromosome Deletion
- Chromosome Disorders/genetics
- Chromosome Duplication/genetics
- Chromosomes, Human, Pair 1/genetics
- Chromosomes, Human, Pair 16/genetics
- Chromosomes, Human, Pair 22/genetics
- Comparative Genomic Hybridization
- DNA Copy Number Variations
- DNA Repair Enzymes/genetics
- DNA-Binding Proteins/genetics
- DiGeorge Syndrome/genetics
- Extracellular Matrix Proteins/genetics
- Female
- Humans
- Hydrolases/genetics
- Intellectual Disability/genetics
- Kruppel-Like Transcription Factors/genetics
- Male
- Megalencephaly/genetics
- Microfilament Proteins/genetics
- Nuclear Proteins/genetics
- Pediatric Obesity/genetics
- Proteins/genetics
- RNA, Messenger/metabolism
- Siblings
- Subcutaneous Fat/metabolism
- Transcription Factors/genetics
- Transcription Factors, General
- Transcriptome
- Young Adult
Collapse
Affiliation(s)
- Maria Pettersson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 77, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Heli Viljakainen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki FI-00029, Finland
| | - Petra Loid
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki FI-00029, Finland
| | - Taina Mustila
- Department of Pediatrics, Seinäjoki Central Hospital, Seinäjoki FI-60100, Finland
| | - Minna Pekkinen
- Folkhälsan Institute of Genetics, Helsinki FI-00290, Finland
| | - Miriam Armenio
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 77, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Johanna C Andersson-Assarsson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
| | - Outi Mäkitie
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 77, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki FI-00029, Finland
- Folkhälsan Institute of Genetics, Helsinki FI-00290, Finland
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 77, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm 171 77, Sweden
| |
Collapse
|
272
|
Zillikens MC, Demissie S, Hsu YH, Yerges-Armstrong LM, Chou WC, Stolk L, Livshits G, Broer L, Johnson T, Koller DL, Kutalik Z, Luan J, Malkin I, Ried JS, Smith AV, Thorleifsson G, Vandenput L, Hua Zhao J, Zhang W, Aghdassi A, Åkesson K, Amin N, Baier LJ, Barroso I, Bennett DA, Bertram L, Biffar R, Bochud M, Boehnke M, Borecki IB, Buchman AS, Byberg L, Campbell H, Campos Obanda N, Cauley JA, Cawthon PM, Cederberg H, Chen Z, Cho NH, Jin Choi H, Claussnitzer M, Collins F, Cummings SR, De Jager PL, Demuth I, Dhonukshe-Rutten RAM, Diatchenko L, Eiriksdottir G, Enneman AW, Erdos M, Eriksson JG, Eriksson J, Estrada K, Evans DS, Feitosa MF, Fu M, Garcia M, Gieger C, Girke T, Glazer NL, Grallert H, Grewal J, Han BG, Hanson RL, Hayward C, Hofman A, Hoffman EP, Homuth G, Hsueh WC, Hubal MJ, Hubbard A, Huffman KM, Husted LB, Illig T, Ingelsson E, Ittermann T, Jansson JO, Jordan JM, Jula A, Karlsson M, Khaw KT, Kilpeläinen TO, Klopp N, Kloth JSL, Koistinen HA, Kraus WE, Kritchevsky S, Kuulasmaa T, Kuusisto J, Laakso M, Lahti J, Lang T, Langdahl BL, Launer LJ, Lee JY, Lerch MM, Lewis JR, Lind L, Lindgren C, Liu Y, et alZillikens MC, Demissie S, Hsu YH, Yerges-Armstrong LM, Chou WC, Stolk L, Livshits G, Broer L, Johnson T, Koller DL, Kutalik Z, Luan J, Malkin I, Ried JS, Smith AV, Thorleifsson G, Vandenput L, Hua Zhao J, Zhang W, Aghdassi A, Åkesson K, Amin N, Baier LJ, Barroso I, Bennett DA, Bertram L, Biffar R, Bochud M, Boehnke M, Borecki IB, Buchman AS, Byberg L, Campbell H, Campos Obanda N, Cauley JA, Cawthon PM, Cederberg H, Chen Z, Cho NH, Jin Choi H, Claussnitzer M, Collins F, Cummings SR, De Jager PL, Demuth I, Dhonukshe-Rutten RAM, Diatchenko L, Eiriksdottir G, Enneman AW, Erdos M, Eriksson JG, Eriksson J, Estrada K, Evans DS, Feitosa MF, Fu M, Garcia M, Gieger C, Girke T, Glazer NL, Grallert H, Grewal J, Han BG, Hanson RL, Hayward C, Hofman A, Hoffman EP, Homuth G, Hsueh WC, Hubal MJ, Hubbard A, Huffman KM, Husted LB, Illig T, Ingelsson E, Ittermann T, Jansson JO, Jordan JM, Jula A, Karlsson M, Khaw KT, Kilpeläinen TO, Klopp N, Kloth JSL, Koistinen HA, Kraus WE, Kritchevsky S, Kuulasmaa T, Kuusisto J, Laakso M, Lahti J, Lang T, Langdahl BL, Launer LJ, Lee JY, Lerch MM, Lewis JR, Lind L, Lindgren C, Liu Y, Liu T, Liu Y, Ljunggren Ö, Lorentzon M, Luben RN, Maixner W, McGuigan FE, Medina-Gomez C, Meitinger T, Melhus H, Mellström D, Melov S, Michaëlsson K, Mitchell BD, Morris AP, Mosekilde L, Newman A, Nielson CM, O'Connell JR, Oostra BA, Orwoll ES, Palotie A, Parker SCJ, Peacock M, Perola M, Peters A, Polasek O, Prince RL, Räikkönen K, Ralston SH, Ripatti S, Robbins JA, Rotter JI, Rudan I, Salomaa V, Satterfield S, Schadt EE, Schipf S, Scott L, Sehmi J, Shen J, Soo Shin C, Sigurdsson G, Smith S, Soranzo N, Stančáková A, Steinhagen-Thiessen E, Streeten EA, Styrkarsdottir U, Swart KMA, Tan ST, Tarnopolsky MA, Thompson P, Thomson CA, Thorsteinsdottir U, Tikkanen E, Tranah GJ, Tuomilehto J, van Schoor NM, Verma A, Vollenweider P, Völzke H, Wactawski-Wende J, Walker M, Weedon MN, Welch R, Wichmann HE, Widen E, Williams FMK, Wilson JF, Wright NC, Xie W, Yu L, Zhou Y, Chambers JC, Döring A, van Duijn CM, Econs MJ, Gudnason V, Kooner JS, Psaty BM, Spector TD, Stefansson K, Rivadeneira F, Uitterlinden AG, Wareham NJ, Ossowski V, Waterworth D, Loos RJF, Karasik D, Harris TB, Ohlsson C, Kiel DP. Large meta-analysis of genome-wide association studies identifies five loci for lean body mass. Nat Commun 2017; 8:80. [PMID: 28724990 PMCID: PMC5517526 DOI: 10.1038/s41467-017-00031-7] [Show More Authors] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 05/02/2017] [Indexed: 12/25/2022] Open
Abstract
Lean body mass, consisting mostly of skeletal muscle, is important for healthy aging. We performed a genome-wide association study for whole body (20 cohorts of European ancestry with n = 38,292) and appendicular (arms and legs) lean body mass (n = 28,330) measured using dual energy X-ray absorptiometry or bioelectrical impedance analysis, adjusted for sex, age, height, and fat mass. Twenty-one single-nucleotide polymorphisms were significantly associated with lean body mass either genome wide (p < 5 × 10-8) or suggestively genome wide (p < 2.3 × 10-6). Replication in 63,475 (47,227 of European ancestry) individuals from 33 cohorts for whole body lean body mass and in 45,090 (42,360 of European ancestry) subjects from 25 cohorts for appendicular lean body mass was successful for five single-nucleotide polymorphisms in/near HSD17B11, VCAN, ADAMTSL3, IRS1, and FTO for total lean body mass and for three single-nucleotide polymorphisms in/near VCAN, ADAMTSL3, and IRS1 for appendicular lean body mass. Our findings provide new insight into the genetics of lean body mass.Lean body mass is a highly heritable trait and is associated with various health conditions. Here, Kiel and colleagues perform a meta-analysis of genome-wide association studies for whole body lean body mass and find five novel genetic loci to be significantly associated.
Collapse
Affiliation(s)
- M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
| | - Serkalem Demissie
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Yi-Hsiang Hsu
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Laura M Yerges-Armstrong
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wen-Chi Chou
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute, Cambridge, MA, 02142, USA
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
| | - Gregory Livshits
- Sackler Faculty of Medicine, Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, 6997801, Israel
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - Linda Broer
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Toby Johnson
- Department of Medical Genetics, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Daniel L Koller
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Ida Malkin
- Sackler Faculty of Medicine, Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Janina S Ried
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | - Liesbeth Vandenput
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Weihua Zhang
- Department Epidemiology and Biostatistics, School of Public Health, Imperial College, London, SW7 2AZ, UK
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
| | - Ali Aghdassi
- Department of Medicine A, University of Greifswald, Greifswald, 17489, Germany
| | - Kristina Åkesson
- Department of Clinical Sciences, Lund University, Malmö, 22362, Sweden
- Department of Orthopedics, Skåne University Hospital, Malmö, S-205 02, Sweden
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
- NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge Metabolic Research Laboratories, Cambridge, CB2 OQQ, UK
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Experimental & Integrative Genomics, University of Lübeck, Lübeck, 23562, Germany
- School of Public Health, Faculty of Medicine, Imperial College London, London, W6 8RP, UK
| | - Rainer Biffar
- Centre of Oral Health, Department of Prosthetic Dentistry, Gerodontology and Biomaterials, University of Greifswald, Greifswald, 17489, Germany
| | - Murielle Bochud
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA
- Division of Biostatistics, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Liisa Byberg
- Department of Surgical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
| | | | - Jane A Cauley
- Department of Epidemiology Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Peggy M Cawthon
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Henna Cederberg
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Zhao Chen
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, 85714, USA
| | - Nam H Cho
- Department of Preventive Medicine, Ajou University School of Medicine, Youngtong-Gu, Suwon, 16499, Korea
| | - Hyung Jin Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju Si, Korea
| | - Melina Claussnitzer
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute, Cambridge, MA, 02142, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, 02139, USA
- Institute of Human Genetics, MRI, Technische Universität München, Munich, 81675, Germany
- Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Francis Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, 20892, USA
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Philip L De Jager
- Harvard Medical School, Boston, MA, 02115, USA
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA
| | - Ilja Demuth
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 13353, Germany
- Institute of Medical and Human Genetics, Charité - Universitätsmedizin Berlin, Berlin, 13353, Germany
| | | | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, H3A 0G1, Canada
- Regional Center for Neurosensory Disorders, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Anke W Enneman
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Mike Erdos
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, 20892, USA
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, 00014, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, 00014, Finland
- Folkhalsan Research Centre, Helsinki, 00250, Finland
- Vasa Central Hospital, Vasa, 65130, Finland
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Joel Eriksson
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Karol Estrada
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Mao Fu
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Melissa Garcia
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Christian Gieger
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Thomas Girke
- Institute for Integrative Genome Biology, University of California, Riverside, CA, 92521, USA
- Department of Botany and Plant Sciences, University of California, Riverside, CA, 92521, USA
| | - Nicole L Glazer
- Departments of Medicine and Epidemiology, Boston University School of Medicine and Public Health, Boston, MA, 02118, USA
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Department of Botany and Plant Sciences, University of California, Riverside, CA, 92521, USA
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- CCG Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, 85764, Germany
- CCG Nutrigenomics and Type 2 Diabetes. Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Jagvir Grewal
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Bok-Ghee Han
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, 28159, Korea
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland, EH4 2XU, UK
| | - Albert Hofman
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Eric P Hoffman
- Department of Pharmaceutical Sciences, SUNY Binghamton, Binghamton, NY, 13902, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, 17487, Germany
| | - Wen-Chi Hsueh
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Monica J Hubal
- Department of Exercise and Nutrition Sciences, George Washington University, Washington, DC, 20052, USA
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20052, USA
| | - Alan Hubbard
- Division of Biostatistics, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Kim M Huffman
- Division of Rheumatology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Lise B Husted
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Department of Human Genetics, Hannover Medical School, Hannover, 30625, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, 30625, Germany
| | - Erik Ingelsson
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Till Ittermann
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE 405 30, Sweden
| | - Joanne M Jordan
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Antti Jula
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Magnus Karlsson
- Department of Clinical Sciences and Orthopaedics, Lund University, Skåne University Hospital SUS, Malmö, 22362, Sweden
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Tuomas O Kilpeläinen
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen, 2100, Denmark
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Norman Klopp
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, 30625, Germany
| | | | - Heikki A Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, 00029, Finland
- Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, 00029, Finland
- Department of Health, National Institute for Health and Welfare, Helsinki, 00271, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, 00290, Finland
| | - William E Kraus
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Stephen Kritchevsky
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Teemu Kuulasmaa
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, FI00014, Finland
| | - Thomas Lang
- University of California San Francisco, San Francisco, CA, 94143, USA
| | - Bente L Langdahl
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Jong-Young Lee
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, 28159, Korea
| | - Markus M Lerch
- Department of Medicine A, University of Greifswald, Greifswald, 17489, Germany
| | - Joshua R Lewis
- School of Medicine and Pharmacology, University of Western Australia, Perth, 6009, Australia
- Centre for Kidney Research, School of Public Health, University of Sydney, Sydney, 2006, Australia
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, OX3 7BN, UK
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, 27517, USA
| | - Tian Liu
- Max Planck Institute for Molecular Genetics, Berlin, 14195, Germany
- Max Planck Institute for Human Development, Berlin, 14195, Germany
| | - Youfang Liu
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Mattias Lorentzon
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Robert N Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - William Maixner
- Regional Center for Neurosensory Disorders, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Fiona E McGuigan
- Department of Clinical Sciences, Lund University, Malmö, 22362, Sweden
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Thomas Meitinger
- Institute of Human Genetics, MRI, Technische Universität München, Munich, 81675, Germany
- Institute of Human Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Håkan Melhus
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Dan Mellström
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, LA, CA, 90089, USA
| | - Karl Michaëlsson
- Department of Surgical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Braxton D Mitchell
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, OX3 7BN, UK
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Leif Mosekilde
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Anne Newman
- Center for Aging and Population Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - Jeffrey R O'Connell
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ben A Oostra
- Department of Clinical Genetics, Erasmus MC, Rotterdam, 300 CA, The Netherlands
- Centre for Medical Systems Biology and Netherlands Consortium on Healthy Aging, Leiden, RC2300, The Netherlands
| | - Eric S Orwoll
- Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, FI00014, Finland
| | - Stephen C J Parker
- Human Genetics and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Munro Peacock
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, FI00014, Finland
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Ozren Polasek
- Faculty of Medicine, Department of Public Health, University of Split, Split, 21000, Croatia
| | - Richard L Prince
- School of Medicine and Pharmacology, University of Western Australia, Perth, 6009, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gardiner Hospital, Perth, 6009, Australia
| | - Katri Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, FI00014, Finland
| | - Stuart H Ralston
- Molecular Medicine Centre, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, EH4 2XU, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Hjelt Institute, University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - John A Robbins
- Department of Medicine, University of California at Davis, Sacramento, CA, 95817, USA
| | - Jerome I Rotter
- Institute for Translational Genomic and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor UCLA Medical Center, Torrance, CA, 90502, USA
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Suzanne Satterfield
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Science, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sabine Schipf
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - Laura Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joban Sehmi
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Jian Shen
- Oregon Health & Science University, Portland, OR, 97239, USA
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Gunnar Sigurdsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Endocrinology and Metabolism, Landspitali, The National University Hospital of Iceland, Reykjavik, 101, Iceland
| | - Shad Smith
- Center for Translational Pain Medicine, Department of Anesthiology, Duke University Medical Center, Durham, NC, 27110, USA
| | - Nicole Soranzo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Elisabeth Steinhagen-Thiessen
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 13353, Germany
| | - Elizabeth A Streeten
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Geriatric Research and Education Clinical Center (GRECC) - Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | | | - Karin M A Swart
- Department of Epidemiology and Biostatistics, and the EMGO Institute, VU University Medical Center, Amsterdam, BT1081, The Netherlands
| | - Sian-Tsung Tan
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Mark A Tarnopolsky
- Department of Medicine, McMaster University Medical Center, Hamilton, ON, Canada, L8N 3Z5
| | - Patricia Thompson
- Department of Pathology, Stony Brook School of Medicine, Stony Brook, NY, 11794, USA
| | - Cynthia A Thomson
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, 85714, USA
| | - Unnur Thorsteinsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- deCODE Genetics, Reykjavik, 101, Iceland
| | - Emmi Tikkanen
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Molecular Medicine Centre, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, EH4 2XU, UK
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Jaakko Tuomilehto
- Vasa Central Hospital, Vasa, 65130, Finland
- Department of Neuroscience and Preventive Medicine, Danube-University Krems, Krems, 3500, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah, 12589, Saudi Arabia
- Dasman Diabetes Institute, Dasman, 15462, Kuwait
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, and the EMGO Institute, VU University Medical Center, Amsterdam, BT1081, The Netherlands
| | - Arjun Verma
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
| | - Peter Vollenweider
- Department of Medicine and Internal Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, CH-1011, Switzerland
| | - Henry Völzke
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - H-Erich Wichmann
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, 81377, Germany
- Institute of Medical Statistics and Epidemiology, Technical University, Munich, 81675, Germany
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - James F Wilson
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland, EH4 2XU, UK
| | - Nicole C Wright
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Weijia Xie
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - John C Chambers
- Department Epidemiology and Biostatistics, School of Public Health, Imperial College, London, SW7 2AZ, UK
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College, London, SW3 6NP, UK
- Imperial College Healthcare NHS Trust, London, W2 1NY, UK
| | - Angela Döring
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
- Centre for Medical Systems Biology and Netherlands Consortium on Healthy Aging, Leiden, RC2300, The Netherlands
| | - Michael J Econs
- Department of Medicine and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Jaspal S Kooner
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
- Imperial College Healthcare NHS Trust, London, W2 1NY, UK
| | - Bruce M Psaty
- Departments of Medicine, Epidemiology, and Health Services, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, 98101, USA
- Kaiser Permanente Washington Health Research Institute, Washington, Seattle, WA, 98101, USA
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - Kari Stefansson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- deCODE Genetics, Reykjavik, 101, Iceland
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Vicky Ossowski
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Dawn Waterworth
- Medical Genetics, GlaxoSmithKline, Philadelphia, PA, 19112, USA
| | - Ruth J F Loos
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute of Child Health and Development, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Genetics of Obesity and Related Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David Karasik
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, 1311502, Israel
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Claes Ohlsson
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Douglas P Kiel
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
- Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
| |
Collapse
|
273
|
Loss of type 9 adenylyl cyclase triggers reduced phosphorylation of Hsp20 and diastolic dysfunction. Sci Rep 2017; 7:5522. [PMID: 28717248 PMCID: PMC5514062 DOI: 10.1038/s41598-017-05816-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/05/2017] [Indexed: 01/16/2023] Open
Abstract
Adenylyl cyclase type 9 (AC9) is found tightly associated with the scaffolding protein Yotiao and the IKs ion channel in heart. But apart from potential IKs regulation, physiological roles for AC9 are unknown. We show that loss of AC9 in mice reduces less than 3% of total AC activity in heart but eliminates Yotiao-associated AC activity. AC9−/− mice exhibit no structural abnormalities but show a significant bradycardia, consistent with AC9 expression in sinoatrial node. Global changes in PKA phosphorylation patterns are not altered in AC9−/− heart, however, basal phosphorylation of heat shock protein 20 (Hsp20) is significantly decreased. Hsp20 binds AC9 in a Yotiao-independent manner and deletion of AC9 decreases Hsp20-associated AC activity in heart. In addition, expression of catalytically inactive AC9 in neonatal cardiomyocytes decreases isoproterenol-stimulated Hsp20 phosphorylation, consistent with an AC9-Hsp20 complex. Phosphorylation of Hsp20 occurs largely in ventricles and is vital for the cardioprotective effects of Hsp20. Decreased Hsp20 phosphorylation suggests a potential baseline ventricular defect for AC9−/−. Doppler echocardiography of AC9−/− displays a decrease in the early ventricular filling velocity and ventricular filling ratio (E/A), indicative of grade 1 diastolic dysfunction and emphasizing the importance of local cAMP production in the context of macromolecular complexes.
Collapse
|
274
|
A global evolutionary and metabolic analysis of human obesity gene risk variants. Gene 2017; 627:412-419. [PMID: 28687331 DOI: 10.1016/j.gene.2017.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/02/2017] [Indexed: 01/20/2023]
Abstract
It is generally accepted that the selection of gene variants during human evolution optimized energy metabolism that now interacts with our obesogenic environment to increase the prevalence of obesity. The purpose of this study was to perform a global evolutionary and metabolic analysis of human obesity gene risk variants (110 human obesity genes with 127 nearest gene risk variants) identified using genome-wide association studies (GWAS) to enhance our knowledge of early and late genotypes. As a result of determining the mean frequency of these obesity gene risk variants in 13 available populations from around the world our results provide evidence for the early selection of ancestral risk variants (defined as selection before migration from Africa) and late selection of derived risk variants (defined as selection after migration from Africa). Our results also provide novel information for association of these obesity genes or encoded proteins with diverse metabolic pathways and other human diseases. The overall results indicate a significant differential evolutionary pattern for the selection of obesity gene ancestral and derived risk variants proposed to optimize energy metabolism in varying global environments and complex association with metabolic pathways and other human diseases. These results are consistent with obesity genes that encode proteins possessing a fundamental role in maintaining energy metabolism and survival during the course of human evolution.
Collapse
|
275
|
Sniekers S, Stringer S, Watanabe K, Jansen PR, Coleman JRI, Krapohl E, Taskesen E, Hammerschlag AR, Okbay A, Zabaneh D, Amin N, Breen G, Cesarini D, Chabris CF, Iacono WG, Ikram MA, Johannesson M, Koellinger P, Lee JJ, Magnusson PKE, McGue M, Miller MB, Ollier WER, Payton A, Pendleton N, Plomin R, Rietveld CA, Tiemeier H, van Duijn CM, Posthuma D. Genome-wide association meta-analysis of 78,308 individuals identifies new loci and genes influencing human intelligence. Nat Genet 2017; 49:1107-1112. [PMID: 28530673 PMCID: PMC5665562 DOI: 10.1038/ng.3869] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/24/2017] [Indexed: 12/12/2022]
Abstract
Intelligence is associated with important economic and health-related life outcomes. Despite intelligence having substantial heritability (0.54) and a confirmed polygenic nature, initial genetic studies were mostly underpowered. Here we report a meta-analysis for intelligence of 78,308 individuals. We identify 336 associated SNPs (METAL P < 5 × 10-8) in 18 genomic loci, of which 15 are new. Around half of the SNPs are located inside a gene, implicating 22 genes, of which 11 are new findings. Gene-based analyses identified an additional 30 genes (MAGMA P < 2.73 × 10-6), of which all but one had not been implicated previously. We show that the identified genes are predominantly expressed in brain tissue, and pathway analysis indicates the involvement of genes regulating cell development (MAGMA competitive P = 3.5 × 10-6). Despite the well-known difference in twin-based heritability for intelligence in childhood (0.45) and adulthood (0.80), we show substantial genetic correlation (rg = 0.89, LD score regression P = 5.4 × 10-29). These findings provide new insight into the genetic architecture of intelligence.
Collapse
Affiliation(s)
- Suzanne Sniekers
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Sven Stringer
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Kyoko Watanabe
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Philip R Jansen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, 3000 CB, Rotterdam, The Netherlands
| | - Jonathan RI Coleman
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK
- NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Trust, London, UK
| | - Eva Krapohl
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK
| | - Erdogan Taskesen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
- Alzheimer Centrum, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Anke R Hammerschlag
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Aysu Okbay
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
- Erasmus University Rotterdam Institute for Behavior and Biology, 3062 PA Rotterdam, The Netherlands
| | - Delilah Zabaneh
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Gerome Breen
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK
- NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Trust, London, UK
| | - David Cesarini
- Center for Experimental Social Science, Department of Economics, New York University, New York, NY 10012
| | - Christopher F Chabris
- Department of Psychology, Union College, Schenectady, NY 12308 (currently at: Geisinger Health System, Danville, PA 17822)
| | - William G Iacono
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455-0344
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, 3000 CB, Rotterdam, The Netherlands
| | - Magnus Johannesson
- Department of Economics, Stockholm School of Economics, 113 83 Stockholm, Sweden
| | - Philipp Koellinger
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
- Erasmus University Rotterdam Institute for Behavior and Biology, 3062 PA Rotterdam, The Netherlands
| | - James J Lee
- Department of Psychology, Harvard University, Cambridge, MA 02138; Department of Psychology, University of Minnesota, Minneapolis, MN 55455-0344
| | - Patrik K. E. Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Matt McGue
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455-0344
| | - Mike B. Miller
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455-0344
| | - William ER Ollier
- Centre for Epidemiology, Division of Population Health, Health Services Research & Primary Care, The University of Manchester
| | - Antony Payton
- Centre for Epidemiology, Division of Population Health, Health Services Research & Primary Care, The University of Manchester
| | - Neil Pendleton
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL
| | - Robert Plomin
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK
| | - Cornelius A Rietveld
- Erasmus University Rotterdam Institute for Behavior and Biology, 3062 PA Rotterdam, The Netherlands
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, 3062 PA Rotterdam, The Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, 3000 CB, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CB, Rotterdam, The Netherlands
- Department of Psychiatry, Erasmus Medical Center, 3000 CB, Rotterdam, The Netherlands
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Translational Epidemiology, Faculty Science, Leiden University, The Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
276
|
Saleheen D, Haycock PC, Zhao W, Rasheed A, Taleb A, Imran A, Abbas S, Majeed F, Akhtar S, Qamar N, Zaman KS, Yaqoob Z, Saghir T, Rizvi SNH, Memon A, Mallick NH, Ishaq M, Rasheed SZ, Memon FUR, Mahmood K, Ahmed N, Frossard P, Tsimikas S, Witztum JL, Marcovina S, Sandhu M, Rader DJ, Danesh J. Apolipoprotein(a) isoform size, lipoprotein(a) concentration, and coronary artery disease: a mendelian randomisation analysis. Lancet Diabetes Endocrinol 2017; 5:524-533. [PMID: 28408323 PMCID: PMC5483508 DOI: 10.1016/s2213-8587(17)30088-8] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND The lipoprotein(a) pathway is a causal factor in coronary heart disease. We used a genetic approach to distinguish the relevance of two distinct components of this pathway, apolipoprotein(a) isoform size and circulating lipoprotein(a) concentration, to coronary heart disease. METHODS In this mendelian randomisation study, we measured lipoprotein(a) concentration and determined apolipoprotein(a) isoform size with a genetic method (kringle IV type 2 [KIV2] repeats in the LPA gene) and a serum-based electrophoretic assay in patients and controls (frequency matched for age and sex) from the Pakistan Risk of Myocardial Infarction Study (PROMIS). We calculated odds ratios (ORs) for myocardial infarction per 1-SD difference in either LPA KIV2 repeats or lipoprotein(a) concentration. In a genome-wide analysis of up to 17 503 participants in PROMIS, we identified genetic variants associated with either apolipoprotein(a) isoform size or lipoprotein(a) concentration. Using a mendelian randomisation study design and genetic data on 60 801 patients with coronary heart disease and 123 504 controls from the CARDIoGRAMplusC4D consortium, we calculated ORs for myocardial infarction with variants that produced similar differences in either apolipoprotein(a) isoform size in serum or lipoprotein(a) concentration. Finally, we compared phenotypic versus genotypic ORs to estimate whether apolipoprotein(a) isoform size, lipoprotein(a) concentration, or both were causally associated with coronary heart disease. FINDINGS The PROMIS cohort included 9015 patients with acute myocardial infarction and 8629 matched controls. In participants for whom KIV2 repeat and lipoprotein(a) data were available, the OR for myocardial infarction was 0·93 (95% CI 0·90-0·97; p<0·0001) per 1-SD increment in LPA KIV2 repeats after adjustment for lipoprotein(a) concentration and conventional lipid concentrations. The OR for myocardial infarction was 1·10 (1·05-1·14; p<0·0001) per 1-SD increment in lipoprotein(a) concentration, after adjustment for LPA KIV2 repeats and conventional lipids. Genome-wide analysis identified rs2457564 as a variant associated with smaller apolipoprotein(a) isoform size, but not lipoprotein(a) concentration, and rs3777392 as a variant associated with lipoprotein(a) concentration, but not apolipoprotein(a) isoform size. In 60 801 patients with coronary heart disease and 123 504 controls, OR for myocardial infarction was 0·96 (0·94-0·98; p<0·0001) per 1-SD increment in apolipoprotein(a) protein isoform size in serum due to rs2457564, which was directionally concordant with the OR observed in PROMIS for a similar change. The OR for myocardial infarction was 1·27 (1·07-1·50; p=0·007) per 1-SD increment in lipoprotein(a) concentration due to rs3777392, which was directionally concordant with the OR observed for a similar change in PROMIS. INTERPRETATION Human genetic data suggest that both smaller apolipoprotein(a) isoform size and increased lipoprotein(a) concentration are independent and causal risk factors for coronary heart disease. Lipoprotein(a)-lowering interventions could be preferentially effective in reducing the risk of coronary heart disease in individuals with smaller apolipoprotein(a) isoforms. FUNDING British Heart Foundation, US National Institutes of Health, Fogarty International Center, Wellcome Trust, UK Medical Research Council, UK National Institute for Health Research, and Pfizer.
Collapse
Affiliation(s)
- Danish Saleheen
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Centre for Non-Communicable Diseases, Karachi, Pakistan.
| | - Philip C Haycock
- Medical Research Council (MRC)/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Wei Zhao
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Asif Rasheed
- Centre for Non-Communicable Diseases, Karachi, Pakistan
| | - Adam Taleb
- University of California San Diego, La Jolla, CA, USA
| | - Atif Imran
- Centre for Non-Communicable Diseases, Karachi, Pakistan
| | - Shahid Abbas
- Faisalabad Institute of Cardiology, Faisalabad, Pakistan
| | - Faisal Majeed
- Centre for Non-Communicable Diseases, Karachi, Pakistan
| | - Saba Akhtar
- Centre for Non-Communicable Diseases, Karachi, Pakistan
| | - Nadeem Qamar
- National Institute of Cardiovascular Disorders, Karachi, Pakistan
| | - Khan Shah Zaman
- National Institute of Cardiovascular Disorders, Karachi, Pakistan
| | - Zia Yaqoob
- National Institute of Cardiovascular Disorders, Karachi, Pakistan
| | - Tahir Saghir
- National Institute of Cardiovascular Disorders, Karachi, Pakistan
| | | | - Anis Memon
- National Institute of Cardiovascular Disorders, Karachi, Pakistan
| | | | | | | | | | | | | | | | | | | | - Santica Marcovina
- Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle, WA, USA
| | - Manjinder Sandhu
- Medical Research Council (MRC)/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK; Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Human Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - John Danesh
- Medical Research Council (MRC)/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK; Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK; National Institute for Health Research Blood and Transplant Research Unit, University of Cambridge, Cambridge, UK; Cambridge British Heart Foundation Centre of Excellence, University of Cambridge, Cambridge, UK.
| |
Collapse
|
277
|
Affiliation(s)
- Christopher Gyngell
- Oxford Uehiro Centre for Practical Ethics, Oxford University, Oxford OX1 3PA, UK
| |
Collapse
|
278
|
Miranda-Lora AL, Cruz M, Aguirre-Hernández J, Molina-Díaz M, Gutiérrez J, Flores-Huerta S, Klünder-Klünder M. Exploring single nucleotide polymorphisms previously related to obesity and metabolic traits in pediatric-onset type 2 diabetes. Acta Diabetol 2017; 54:653-662. [PMID: 28401323 DOI: 10.1007/s00592-017-0987-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/27/2017] [Indexed: 12/21/2022]
Abstract
AIMS To evaluate the association of 64 obesity-related polymorphisms with pediatric-onset type 2 diabetes and other glucose- and insulin-related traits in Mexican children. METHODS Case-control and case-sibling designs were followed. We studied 99 patients with pediatric-onset type 2 diabetes, their siblings (n = 101) without diabetes, 83 unrelated pediatric controls and 137 adult controls. Genotypes were determined for 64 single nucleotide polymorphisms, and a possible association was examined between those genotypes and type 2 diabetes and other quantitative traits, after adjusting for age, sex and body mass index. RESULTS In the case-pediatric control and case-adult control analyses, five polymorphisms were associated with increased likelihood of pediatric-onset type 2 diabetes; only one of these polymorphisms (CADM2/rs1307880) also showed a consistent effect in the case-sibling analysis. The associations in the combined analysis were as follows: ADORA1/rs903361 (OR 1.9, 95% CI 1.2; 3.0); CADM2/rs13078807 (OR 2.2, 95% CI 1.2; 4.0); GNPDA2/rs10938397 (OR 2.2, 95% CI 1.4; 3.7); VEGFA/rs6905288 (OR 1.4, 95% CI 1.1; 2.1) and FTO/rs9939609 (OR 1.8, 95% CI 1.0; 3.2). We also identified 16 polymorphisms nominally associated with quantitative traits in participants without diabetes. CONCLUSIONS ADORA/rs903361, CADM2/rs13078807, GNPDA2/rs10938397, VEGFA/rs6905288 and FTO/rs9939609 are associated with an increased risk of pediatric-onset type 2 diabetes in the Mexican population.
Collapse
Affiliation(s)
| | - Miguel Cruz
- Medical Research Unit in Biochemistry, Hospital de Especialidades Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Jesús Aguirre-Hernández
- Laboratory of Genomics, Genetics and Bioinformatics, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Mario Molina-Díaz
- Department of Endocrinology, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Jorge Gutiérrez
- Medical Research Unit in Biochemistry, Hospital de Especialidades Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Samuel Flores-Huerta
- Department of Community Health Research, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Miguel Klünder-Klünder
- Department of Community Health Research, Hospital Infantil de México Federico Gómez, Mexico City, Mexico.
- Research Committee, Latin American Society for Pediatric Gastroenterology, Hepatology and Nutrition (LASPGHAN), Mexico City, Mexico.
| |
Collapse
|
279
|
Effect of dietary consumption as a modifier on the association between FTO gene variants and excess body weight in children from an admixed population in Brazil: the Social Changes, Asthma and Allergy in Latin America (SCAALA) cohort study. Br J Nutr 2017; 117:1503-1510. [PMID: 28659218 DOI: 10.1017/s0007114517001386] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Previous studies have shown associations of variants of the FTO gene with body weight, but none of these have involved Latin American populations with a high level of miscegenation, as is seen in the north-eastern Brazilian population. This study evaluated the association between SNP in the FTO gene and excess weight in Salvador, Bahia, Brazil. In addition, the effect of diet as a modifier on this association was also investigated. This cross-sectional study included 1191 participants aged 4-11 years, who were genotyped for 400 variants of the FTO gene. Direct anthropometric measures were made and dietary data were obtained by 24-h food recall. Multivariate logistic regression analyses were used to assess the associations of interest. Overall, 11·2 % of the individuals included in the study were overweight/obese. Interactions were identified between the percentage energy intake from proteins and obesity risk linked to the rs62048379 SNP (P interaction=0·01) and also between fat intake (PUFA:SFA ratio) and obesity risk linked to the rs62048379 SNP (P interaction=0·01). The T allele for the variant rs62048379 was positively associated with overweight/obesity in individuals whose percentage energy intake from protein was above the median (OR 2·00; 95 % CI 1·05, 3·82). The rs62048379 SNP was also associated with overweight/obesity in individuals whose PUFA:SFA ratio was below the median (OR 1·63; 95 % CI 1·05, 2·55). The association between FTO gene variants and excess body weight can be modulated by dietary characteristics, particularly by fatty acid distribution and dietary protein intake in children.
Collapse
|
280
|
Maddirevula S, AlZahrani F, Anazi S, Almureikhi M, Ben-Omran T, Abdel-Salam GMH, Hashem M, Ibrahim N, Abdulwahab FM, Meriki N, Bashiri FA, Thong MK, Muthukumarasamy P, Azwani Mazlan R, Shaheen R, Alkuraya FS. GWAS signals revisited using human knockouts. Genet Med 2017. [PMID: 28640246 DOI: 10.1038/gim.2017.78] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PurposeGenome-wide association studies (GWAS) have been instrumental to our understanding of the genetic risk determinants of complex traits. A common challenge in GWAS is the interpretation of signals, which are usually attributed to the genes closest to the polymorphic markers that display the strongest statistical association. Naturally occurring complete loss of function (knockout) of these genes in humans can inform GWAS interpretation by unmasking their deficiency state in a clinical context.MethodsWe exploited the unique population structure of Saudi Arabia to identify novel knockout events in genes previously highlighted in GWAS using combined autozygome/exome analysis.ResultsWe report five families with homozygous truncating mutations in genes that had only been linked to human disease through GWAS. The phenotypes observed in the natural knockouts for these genes (TRAF3IP2, FRMD3, RSRC1, BTBD9, and PXDNL) range from consistent with, to unrelated to, the previously reported GWAS phenotype.ConclusionWe expand the role of human knockouts in the medical annotation of the human genome, and show their potential value in informing the interpretation of GWAS of complex traits.
Collapse
Affiliation(s)
- Sateesh Maddirevula
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fatema AlZahrani
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Shams Anazi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mariam Almureikhi
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Tawfeg Ben-Omran
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
| | - Ghada M H Abdel-Salam
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Mais Hashem
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Niema Ibrahim
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Firdous M Abdulwahab
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Neama Meriki
- Department of Obstetrics and Gynecology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fahad A Bashiri
- Department of Pediatrics, College of Medicine & King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia
| | - Meow-Keong Thong
- Genetics and Metabolism Unit, Department of Paediatrics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Premala Muthukumarasamy
- Genetics and Metabolism Unit, Department of Paediatrics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Rifhan Azwani Mazlan
- Genetics and Metabolism Unit, Department of Paediatrics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ranad Shaheen
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| |
Collapse
|
281
|
Rodriguez-Broadbent H, Law PJ, Sud A, Palin K, Tuupanen S, Gylfe A, Hänninen UA, Cajuso T, Tanskanen T, Kondelin J, Kaasinen E, Sarin AP, Ripatti S, Eriksson JG, Rissanen H, Knekt P, Pukkala E, Jousilahti P, Salomaa V, Palotie A, Renkonen-Sinisalo L, Lepistö A, Böhm J, Mecklin JP, Al-Tassan NA, Palles C, Martin L, Barclay E, Farrington SM, Timofeeva MN, Meyer BF, Wakil SM, Campbell H, Smith CG, Idziaszczyk S, Maughan TS, Kaplan R, Kerr R, Kerr D, Passarelli MN, Figueiredo JC, Buchanan DD, Win AK, Hopper JL, Jenkins MA, Lindor NM, Newcomb PA, Gallinger S, Conti D, Schumacher F, Casey G, Aaltonen LA, Cheadle JP, Tomlinson IP, Dunlop MG, Houlston RS. Mendelian randomisation implicates hyperlipidaemia as a risk factor for colorectal cancer. Int J Cancer 2017; 140:2701-2708. [PMID: 28340513 PMCID: PMC6135234 DOI: 10.1002/ijc.30709] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/26/2017] [Accepted: 02/08/2017] [Indexed: 01/07/2023]
Abstract
While elevated blood cholesterol has been associated with an increased risk of colorectal cancer (CRC) in observational studies, causality is uncertain. Here we apply a Mendelian randomisation (MR) analysis to examine the potential causal relationship between lipid traits and CRC risk. We used single nucleotide polymorphisms (SNPs) associated with blood levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) as instrumental variables (IV). We calculated MR estimates for each risk factor with CRC using SNP-CRC associations from 9,254 cases and 18,386 controls. Genetically predicted higher TC was associated with an elevated risk of CRC (odds ratios (OR) per unit SD increase = 1.46, 95% confidence interval [CI]: 1.20-1.79, p = 1.68 × 10-4 ). The pooled ORs for LDL, HDL, and TG were 1.05 (95% CI: 0.92-1.18, p = 0.49), 0.94 (95% CI: 0.84-1.05, p = 0.27), and 0.98 (95% CI: 0.85-1.12, p = 0.75) respectively. A genetic risk score for 3-hydoxy-3-methylglutaryl-coenzyme A reductase (HMGCR) to mimic the effects of statin therapy was associated with a reduced CRC risk (OR = 0.69, 95% CI: 0.49-0.99, p = 0.046). This study supports a causal relationship between higher levels of TC with CRC risk, and a further rationale for implementing public health strategies to reduce the prevalence of hyperlipidaemia.
Collapse
Affiliation(s)
| | - Philip J. Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Kimmo Palin
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Sari Tuupanen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Alexandra Gylfe
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Ulrika A. Hänninen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Tatiana Cajuso
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Tomas Tanskanen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Johanna Kondelin
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Eevi Kaasinen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
- Department of Public Health, University of Helsinki, Helsinki, 00014, Finland
| | - Johan G. Eriksson
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Folkhälsan Research Centre, Helsinki, 00250, Finland
- Unit of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, 00014, Finland
| | - Harri Rissanen
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Paul Knekt
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Eero Pukkala
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, 00130, Finland
- School of Health Sciences, University of Tampere, Tampere, 33014, Finland
| | - Pekka Jousilahti
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Laura Renkonen-Sinisalo
- Abdominal Center, Department of Surgery, Helsinki University Hospital, Helsinki, 00029, Finland
| | - Anna Lepistö
- Abdominal Center, Department of Surgery, Helsinki University Hospital, Helsinki, 00029, Finland
| | - Jan Böhm
- Department of Pathology, Central Finland Central Hospital, Jyväskylä, 40620, Finland
| | - Jukka-Pekka Mecklin
- Department of Surgery, Jyväskylä Central Hospital, University of Eastern Finland, Jyväskylä, 40620, Finland
| | - Nada A. Al-Tassan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 12713, Saudi Arabia
| | - Claire Palles
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Lynn Martin
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Ella Barclay
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Susan M. Farrington
- Colon Cancer Genetics Group, University of Edinburgh and MRC Human Genetics Unit, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Maria N. Timofeeva
- Colon Cancer Genetics Group, University of Edinburgh and MRC Human Genetics Unit, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Brian F. Meyer
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 12713, Saudi Arabia
| | - Salma M. Wakil
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 12713, Saudi Arabia
| | - Harry Campbell
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Christopher G. Smith
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Shelley Idziaszczyk
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Timothy S. Maughan
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Richard Kaplan
- MRC Clinical Trials Unit, Aviation House, London, WC2B 6NH, UK
| | - Rachel Kerr
- Oxford Cancer Centre, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - David Kerr
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Michael N. Passarelli
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Jane C. Figueiredo
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel D. Buchanan
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - Aung K. Win
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - John L. Hopper
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - Mark A. Jenkins
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Victoria, 3010, Australia
| | - Noralane M. Lindor
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Polly A. Newcomb
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - David Conti
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Fred Schumacher
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Graham Casey
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lauri A. Aaltonen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, 00014, Finland
| | - Jeremy P. Cheadle
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Ian P. Tomlinson
- Wellcome Trust Centre for Human Genetics and NIHR Comprehensive Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Malcolm G. Dunlop
- Colon Cancer Genetics Group, University of Edinburgh and MRC Human Genetics Unit, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| |
Collapse
|
282
|
Tachmazidou I, Süveges D, Min JL, Ritchie GRS, Steinberg J, Walter K, Iotchkova V, Schwartzentruber J, Huang J, Memari Y, McCarthy S, Crawford AA, Bombieri C, Cocca M, Farmaki AE, Gaunt TR, Jousilahti P, Kooijman MN, Lehne B, Malerba G, Männistö S, Matchan A, Medina-Gomez C, Metrustry SJ, Nag A, Ntalla I, Paternoster L, Rayner NW, Sala C, Scott WR, Shihab HA, Southam L, St Pourcain B, Traglia M, Trajanoska K, Zaza G, Zhang W, Artigas MS, Bansal N, Benn M, Chen Z, Danecek P, Lin WY, Locke A, Luan J, Manning AK, Mulas A, Sidore C, Tybjaerg-Hansen A, Varbo A, Zoledziewska M, Finan C, Hatzikotoulas K, Hendricks AE, Kemp JP, Moayyeri A, Panoutsopoulou K, Szpak M, Wilson SG, Boehnke M, Cucca F, Di Angelantonio E, Langenberg C, Lindgren C, McCarthy MI, Morris AP, Nordestgaard BG, Scott RA, Tobin MD, Wareham NJ, Burton P, Chambers JC, Smith GD, Dedoussis G, Felix JF, Franco OH, Gambaro G, Gasparini P, Hammond CJ, Hofman A, Jaddoe VWV, Kleber M, Kooner JS, Perola M, Relton C, Ring SM, Rivadeneira F, Salomaa V, Spector TD, Stegle O, Toniolo D, Uitterlinden AG, Barroso I, Greenwood CMT, Perry JRB, et alTachmazidou I, Süveges D, Min JL, Ritchie GRS, Steinberg J, Walter K, Iotchkova V, Schwartzentruber J, Huang J, Memari Y, McCarthy S, Crawford AA, Bombieri C, Cocca M, Farmaki AE, Gaunt TR, Jousilahti P, Kooijman MN, Lehne B, Malerba G, Männistö S, Matchan A, Medina-Gomez C, Metrustry SJ, Nag A, Ntalla I, Paternoster L, Rayner NW, Sala C, Scott WR, Shihab HA, Southam L, St Pourcain B, Traglia M, Trajanoska K, Zaza G, Zhang W, Artigas MS, Bansal N, Benn M, Chen Z, Danecek P, Lin WY, Locke A, Luan J, Manning AK, Mulas A, Sidore C, Tybjaerg-Hansen A, Varbo A, Zoledziewska M, Finan C, Hatzikotoulas K, Hendricks AE, Kemp JP, Moayyeri A, Panoutsopoulou K, Szpak M, Wilson SG, Boehnke M, Cucca F, Di Angelantonio E, Langenberg C, Lindgren C, McCarthy MI, Morris AP, Nordestgaard BG, Scott RA, Tobin MD, Wareham NJ, Burton P, Chambers JC, Smith GD, Dedoussis G, Felix JF, Franco OH, Gambaro G, Gasparini P, Hammond CJ, Hofman A, Jaddoe VWV, Kleber M, Kooner JS, Perola M, Relton C, Ring SM, Rivadeneira F, Salomaa V, Spector TD, Stegle O, Toniolo D, Uitterlinden AG, Barroso I, Greenwood CMT, Perry JRB, Walker BR, Butterworth AS, Xue Y, Durbin R, Small KS, Soranzo N, Timpson NJ, Zeggini E. Whole-Genome Sequencing Coupled to Imputation Discovers Genetic Signals for Anthropometric Traits. Am J Hum Genet 2017; 100:865-884. [PMID: 28552196 PMCID: PMC5473732 DOI: 10.1016/j.ajhg.2017.04.014] [Show More Authors] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/21/2017] [Indexed: 01/05/2023] Open
Abstract
Deep sequence-based imputation can enhance the discovery power of genome-wide association studies by assessing previously unexplored variation across the common- and low-frequency spectra. We applied a hybrid whole-genome sequencing (WGS) and deep imputation approach to examine the broader allelic architecture of 12 anthropometric traits associated with height, body mass, and fat distribution in up to 267,616 individuals. We report 106 genome-wide significant signals that have not been previously identified, including 9 low-frequency variants pointing to functional candidates. Of the 106 signals, 6 are in genomic regions that have not been implicated with related traits before, 28 are independent signals at previously reported regions, and 72 represent previously reported signals for a different anthropometric trait. 71% of signals reside within genes and fine mapping resolves 23 signals to one or two likely causal variants. We confirm genetic overlap between human monogenic and polygenic anthropometric traits and find signal enrichment in cis expression QTLs in relevant tissues. Our results highlight the potential of WGS strategies to enhance biologically relevant discoveries across the frequency spectrum.
Collapse
Affiliation(s)
- Ioanna Tachmazidou
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Dániel Süveges
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Josine L Min
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Graham R S Ritchie
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Usher Institute of Population Health Sciences & Informatics, University of Edinburgh, Edinburgh EH16 4UX, UK; MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH16 4UX, UK
| | - Julia Steinberg
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Klaudia Walter
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Valentina Iotchkova
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SD, UK
| | | | - Jie Huang
- Boston VA Research Institute, Boston, MA 02130, USA
| | - Yasin Memari
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Shane McCarthy
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Andrew A Crawford
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK; BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Cristina Bombieri
- Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Massimiliano Cocca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34100, Italy
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens 17671, Greece
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Pekka Jousilahti
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Marjolein N Kooijman
- The Generation R Study Group, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Pediatrics, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Giovanni Malerba
- Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Satu Männistö
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Angela Matchan
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Sarah J Metrustry
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Abhishek Nag
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Nigel W Rayner
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - William R Scott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK
| | - Hashem A Shihab
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Lorraine Southam
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK; Max Planck Institute for Psycholinguistics, Nijmegen 6500, the Netherlands
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Katerina Trajanoska
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Gialuigi Zaza
- Renal Unit, Department of Medicine, Verona University Hospital, Verona 37126, Italy
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK
| | - María S Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Narinder Bansal
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Marianne Benn
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Zhongsheng Chen
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Petr Danecek
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Wei-Yu Lin
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Adam Locke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Alisa K Manning
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Harvard University Medical School, Boston, MA 02115, USA
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR), Cagliari 09100, Italy; Università degli Studi di Sassari, Sassari 07100, Italy
| | - Carlo Sidore
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR), Cagliari 09100, Italy
| | - Anne Tybjaerg-Hansen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Anette Varbo
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | | | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, UK
| | | | - Audrey E Hendricks
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA
| | - John P Kemp
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK; University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD 4072, Australia
| | - Alireza Moayyeri
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK; Institute of Health Informatics, University College London, London NW1 2DA, UK
| | | | - Michal Szpak
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Scott G Wilson
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK; School of Medicine and Pharmacology, The University of Western Australia, Crawley, WA 6009, Australia; Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR), Cagliari 09100, Italy; Università degli Studi di Sassari, Sassari 07100, Italy
| | - Emanuele Di Angelantonio
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge CB1 8RN, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Li Ka Shing Centre for Health Information and Discovery, The Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK; Estonian Genome Center, University of Tartu, Tartu, Tartumaa 51010, Estonia
| | - Børge G Nordestgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Martin D Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK; National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | | | - Paul Burton
- D2K Research Group, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK; Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens 17671, Greece
| | - Janine F Felix
- The Generation R Study Group, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Pediatrics, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Giovanni Gambaro
- Division of Nephrology and Dialysis, Columbus-Gemelli University Hospital, Catholic University, Rome 00168, Italy
| | - Paolo Gasparini
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34100, Italy; Medical Genetics, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste 34100, Italy
| | - Christopher J Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Pediatrics, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Marcus Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK; Imperial College Healthcare NHS Trust, London W2 1NY, UK; National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Markus Perola
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland; Estonian Genome Center, University of Tartu, Tartu, Tartumaa 51010, Estonia; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki 00290, Finland
| | - Caroline Relton
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Susan M Ring
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Veikko Salomaa
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SD, UK
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | | | | | | | - Inês Barroso
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; University of Cambridge Metabolic Research Laboratories, and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Celia M T Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T 1E2, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC H3A 1A2, Canada; Department of Oncology, McGill University, Montréal, QC H2W 1S6, Canada
| | - John R B Perry
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Adam S Butterworth
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge CB1 8RN, UK
| | - Yali Xue
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Richard Durbin
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Nicole Soranzo
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge CB1 8RN, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AH, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Eleftheria Zeggini
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK.
| |
Collapse
|
283
|
Hebbar P, Alkayal F, Nizam R, Melhem M, Elkum N, John SE, Abufarha M, Alsmadi O, Thanaraj TA. The TCN2 variant of rs9606756 [Ile23Val] acts as risk loci for obesity-related traits and mediates by interacting with Apo-A1. Obesity (Silver Spring) 2017; 25:1098-1108. [PMID: 28417558 DOI: 10.1002/oby.21826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/02/2017] [Accepted: 02/22/2017] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Despite alarming obesity levels in the Arabian Peninsula, its population lacks convincingly identified genetic determinants of obesity. A genome-wide association study was performed for obesity-related anthropometric traits in Arabs and to decipher mechanisms by which the variants mediate traits. METHODS The Illumina HumanOmniExpress BeadChip was used to genotype 1,353 Arab individuals (largely with Class I obesity) from Kuwait. Genome-wide association tests for obesity-related anthropometric traits were performed. Top associations were tested for replication in an independent cohort (1,176 unrelated Arabs). Resultant variants were investigated for interactions with obesity-related plasma biomarkers. Pathway analysis was performed on genes harboring markers in linkage disequilibrium (LD) with identified variants. RESULTS The rs9606756[c.67A>G,p.Ile23Val] variant from TCN2 was associated with waist circumference (WC) at nearly genome-wide significance (P = 8.92E-08). WC was inversely related with Apo-A1 or high-density lipoprotein levels; individuals with the AG genotype exhibited stronger relationship than those with the reference AA genotype. Interaction involving the AG genotype (effect allele = G) significantly contributed to an increase in anthropometric traits (particularly WC). Genes harboring single-nucleotide polymorphisms in LD with rs9606756 mapped onto an interaction network (with TP53 as central element) of established obesity/diabetes-related protein components. CONCLUSIONS The TCN2 variant acts as a risk factor for WC in the Arab population. The variant mediates obesity-related anthropometric traits via interactions with Apo-A1/high-density lipoprotein or TP53.
Collapse
Affiliation(s)
| | | | | | | | - Naser Elkum
- Sidra Medical and Research Center, Research Department Doha, Qatar
| | | | | | | | | |
Collapse
|
284
|
Fernández-Rhodes L, Gong J, Haessler J, Franceschini N, Graff M, Nishimura KK, Wang Y, Highland HM, Yoneyama S, Bush WS, Goodloe R, Ritchie MD, Crawford D, Gross M, Fornage M, Buzkova P, Tao R, Isasi C, Avilés-Santa L, Daviglus M, Mackey RH, Houston D, Gu CC, Ehret G, Nguyen KDH, Lewis CE, Leppert M, Irvin MR, Lim U, Haiman CA, Le Marchand L, Schumacher F, Wilkens L, Lu Y, Bottinger EP, Loos RJL, Sheu WHH, Guo X, Lee WJ, Hai Y, Hung YJ, Absher D, Wu IC, Taylor KD, Lee IT, Liu Y, Wang TD, Quertermous T, Juang JMJ, Rotter JI, Assimes T, Hsiung CA, Chen YDI, Prentice R, Kuller LH, Manson JE, Kooperberg C, Smokowski P, Robinson WR, Gordon-Larsen P, Li R, Hindorff L, Buyske S, Matise TC, Peters U, North KE. Trans-ethnic fine-mapping of genetic loci for body mass index in the diverse ancestral populations of the Population Architecture using Genomics and Epidemiology (PAGE) Study reveals evidence for multiple signals at established loci. Hum Genet 2017; 136:771-800. [PMID: 28391526 PMCID: PMC5485655 DOI: 10.1007/s00439-017-1787-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 03/23/2017] [Indexed: 11/26/2022]
Abstract
Most body mass index (BMI) genetic loci have been identified in studies of primarily European ancestries. The effect of these loci in other racial/ethnic groups is less clear. Thus, we aimed to characterize the generalizability of 170 established BMI variants, or their proxies, to diverse US populations and trans-ethnically fine-map 36 BMI loci using a sample of >102,000 adults of African, Hispanic/Latino, Asian, European and American Indian/Alaskan Native descent from the Population Architecture using Genomics and Epidemiology Study. We performed linear regression of the natural log of BMI (18.5-70 kg/m2) on the additive single nucleotide polymorphisms (SNPs) at BMI loci on the MetaboChip (Illumina, Inc.), adjusting for age, sex, population stratification, study site, or relatedness. We then performed fixed-effect meta-analyses and a Bayesian trans-ethnic meta-analysis to empirically cluster by allele frequency differences. Finally, we approximated conditional and joint associations to test for the presence of secondary signals. We noted directional consistency with the previously reported risk alleles beyond what would have been expected by chance (binomial p < 0.05). Nearly, a quarter of the previously described BMI index SNPs and 29 of 36 densely-genotyped BMI loci on the MetaboChip replicated/generalized in trans-ethnic analyses. We observed multiple signals at nine loci, including the description of seven loci with novel multiple signals. This study supports the generalization of most common genetic loci to diverse ancestral populations and emphasizes the importance of dense multiethnic genomic data in refining the functional variation at genetic loci of interest and describing several loci with multiple underlying genetic variants.
Collapse
Affiliation(s)
- Lindsay Fernández-Rhodes
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jian Gong
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nora Franceschini
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mariaelisa Graff
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine K Nishimura
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yujie Wang
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather M Highland
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sachiko Yoneyama
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William S Bush
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Robert Goodloe
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA
| | - Marylyn D Ritchie
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Dana Crawford
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Myron Gross
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Myriam Fornage
- Center for Human Genetics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Petra Buzkova
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Ran Tao
- Department of Biostatistics, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carmen Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Martha Daviglus
- Insitute of Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Rachel H Mackey
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Denise Houston
- Geriatrics and Gerontology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - C Charles Gu
- Division of Biostatistics, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Georg Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
- Division of Cardiology, Geneva University Hospital, Geneva, OH, Switzerland
| | - Khanh-Dung H Nguyen
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Cora E Lewis
- Department of Medicine, University of Alabama, Birmingham, AL, USA
| | - Mark Leppert
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | | | - Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Fredrick Schumacher
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lynne Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Yingchang Lu
- Charles R. Bronfman Instituted for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erwin P Bottinger
- Charles R. Bronfman Instituted for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruth J L Loos
- Charles R. Bronfman Instituted for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wayne H-H Sheu
- Division of Endocrine and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Defense Medical Center, National Yang-Ming University, Taipei, Taiwan
| | - Xiuqing Guo
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yang Hai
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yi-Jen Hung
- Division of Endocrinology and Metabolism, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - I-Chien Wu
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan Town, Taiwan
| | - Kent D Taylor
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - I-Te Lee
- Division of Endocrine and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Defense Medical Center, National Yang-Ming University, Taipei, Taiwan
| | - Yeheng Liu
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Tzung-Dau Wang
- Division of Cardiology, Department of Internal Medicine, Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Thomas Quertermous
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jyh-Ming J Juang
- Division of Cardiology, Department of Internal Medicine, Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Jerome I Rotter
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Themistocles Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Chao A Hsiung
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan Town, Taiwan
| | - Yii-Der Ida Chen
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ross Prentice
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lewis H Kuller
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - JoAnn E Manson
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Paul Smokowski
- School of Social Welfare, The University of Kansas, Lawrence, KS, USA
| | - Whitney R Robinson
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Penny Gordon-Larsen
- Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rongling Li
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lucia Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven Buyske
- Department of Statistics and Biostatistics, Rutgers University, Piscataway, NJ, USA
| | - Tara C Matise
- Department of Genetics, Rutgers University, Piscataway, NJ, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kari E North
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
285
|
Hong C, Ning Y, Wei P, Cao Y, Chen Y. A semiparametric model for vQTL mapping. Biometrics 2017; 73:571-581. [PMID: 27861717 PMCID: PMC5780188 DOI: 10.1111/biom.12612] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 07/01/2016] [Accepted: 08/01/2016] [Indexed: 11/30/2022]
Abstract
Quantitative trait locus analysis has been used as an important tool to identify markers where the phenotype or quantitative trait is linked with the genotype. Most existing tests for single locus association with quantitative traits aim at the detection of the mean differences across genotypic groups. However, recent research has revealed functional genetic loci that affect the variance of traits, known as variability-controlling quantitative trait locus. In addition, it has been suggested that many genotypes have both mean and variance effects, while the mean effects or variance effects alone may not be strong enough to be detected. The existing methods accounting for unequal variances include the Levene's test, the Lepage test, and the D-test, but suffer from their limitations of lack of robustness or lack of power. We propose a semiparametric model and a novel pairwise conditional likelihood ratio test. Specifically, the semiparametric model is designed to identify the combined differences in higher moments among genotypic groups. The pairwise likelihood is constructed based on conditioning procedure, where the unknown reference distribution is eliminated. We show that the proposed pairwise likelihood ratio test has a simple asymptotic chi-square distribution, which does not require permutation or bootstrap procedures. Simulation studies show that the proposed test performs well in controlling Type I errors and having competitive power in identifying the differences across genotypic groups. In addition, the proposed test has certain robustness to model mis-specifications. The proposed test is illustrated by an example of identifying both mean and variances effects in body mass index using the Framingham Heart Study data.
Collapse
Affiliation(s)
- Chuan Hong
- Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Yang Ning
- Department of Statistical Science, Cornell University, Ithaca, NY 14853, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ying Cao
- Department of Biostatistics, The University of Texas School of Public Health, Houston, TX 77030, USA
| | - Yong Chen
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
286
|
Structural variants caused by Alu insertions are associated with risks for many human diseases. Proc Natl Acad Sci U S A 2017; 114:E3984-E3992. [PMID: 28465436 DOI: 10.1073/pnas.1704117114] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Interspersed repeat sequences comprise much of our DNA, although their functional effects are poorly understood. The most commonly occurring repeat is the Alu short interspersed element. New Alu insertions occur in human populations, and have been responsible for several instances of genetic disease. In this study, we sought to determine if there are instances of polymorphic Alu insertion variants that function in a common variant, common disease paradigm. We cataloged 809 polymorphic Alu elements mapping to 1,159 loci implicated in disease risk by genome-wide association study (GWAS) (P < 10-8). We found that Alu insertion variants occur disproportionately at GWAS loci (P = 0.013). Moreover, we identified 44 of these Alu elements in linkage disequilibrium (r2 > 0.7) with the trait-associated SNP. This figure represents a >20-fold increase in the number of polymorphic Alu elements associated with human phenotypes. This work provides a broader perspective on how structural variants in repetitive DNAs may contribute to human disease.
Collapse
|
287
|
Frau F, Crowther D, Ruetten H, Allebrandt KV. Type-2 diabetes-associated variants with cross-trait relevance: Post-GWAs strategies for biological function interpretation. Mol Genet Metab 2017; 121:43-50. [PMID: 28385534 DOI: 10.1016/j.ymgme.2017.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/13/2017] [Accepted: 03/13/2017] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies (GWAs) for type 2 diabetes (T2D) have been successful in identifying many loci with robust association signals. Nevertheless, there is a clear need for post-GWAs strategies to understand mechanism of action and clinical relevance of these variants. The association of several comorbidities with T2D suggests a common etiology for these phenotypes and complicates the management of the disease. In this study, we focused on the genetics underlying these relationships, using systems genomics to identify genetic variation associated with T2D and 12 other traits. GWAs studies summary statistics for pairwise comparisons were obtained for glycemic traits, obesity, coronary artery disease, and lipids from large consortia GWAs meta-analyses. We used a network medicine approach to leverage experimental information about the identified genes and variants with cross traits effects for biological function interpretation. We identified a set of 38 genetic variants with cross traits effects that point to a main network of genes that should be relevant for T2D and its comorbidities. We prioritized the T2D associated genes based on the number of traits they showed association with and the experimental evidence showing their relation to the disease etiology. In this study, we demonstrated how systems genomics and network medicine approaches can shed light into GWAs discoveries, translating findings into a more therapeutically relevant context.
Collapse
Affiliation(s)
- Francesca Frau
- Department of Translational Informatics, R&D Translational Med. and Early Development, Sanofi-Aventis, Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Daniel Crowther
- Department of Translational Informatics, R&D Translational Med. and Early Development, Sanofi-Aventis, Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Hartmut Ruetten
- R&D Translational Med. and Early Clinical, Sanofi-Aventis, Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Karla V Allebrandt
- Department of Translational Informatics, R&D Translational Med. and Early Development, Sanofi-Aventis, Deutschland GmbH, Industriepark Höchst, D-65926 Frankfurt am Main, Germany.
| |
Collapse
|
288
|
Fall T, Mendelson M, Speliotes EK. Recent Advances in Human Genetics and Epigenetics of Adiposity: Pathway to Precision Medicine? Gastroenterology 2017; 152:1695-1706. [PMID: 28214526 PMCID: PMC5576453 DOI: 10.1053/j.gastro.2017.01.054] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 12/26/2022]
Abstract
Obesity is a heritable trait that contributes to substantial global morbidity and mortality. Here, we summarize findings from the past decade of genetic and epigenetic research focused on unravelling the underpinnings of adiposity. More than 140 genetic regions now are known to influence adiposity traits. The genetics of general adiposity, as measured by body mass index, and that of abdominal obesity, as measured by waist-to-hip ratio, have distinct biological backgrounds. Gene expression associated with general adiposity is enriched in the nervous system. In contrast, genes associated with abdominal adiposity function in adipose tissue. Recent population-based epigenetic analyses have highlighted additional distinct loci. We discuss how associated genetic variants can lead to understanding causal mechanisms, and to disentangling reverse causation in epigenetic analyses. Discoveries emerging from population genomics are identifying new disease markers and potential novel drug targets to better define and combat obesity and related diseases.
Collapse
Affiliation(s)
- Tove Fall
- Department of Medical Sciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael Mendelson
- The Framingham Heart Study, Framingham, Massachusetts,Population Sciences Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland,Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
| | - Elizabeth K. Speliotes
- Department of Internal Medicine, Division of Gastroenterology,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
289
|
Mahendran Y, Jonsson A, Have CT, Allin KH, Witte DR, Jørgensen ME, Grarup N, Pedersen O, Kilpeläinen TO, Hansen T. Genetic evidence of a causal effect of insulin resistance on branched-chain amino acid levels. Diabetologia 2017; 60:873-878. [PMID: 28184960 DOI: 10.1007/s00125-017-4222-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/25/2017] [Indexed: 01/21/2023]
Abstract
AIMS/HYPOTHESIS Fasting plasma levels of branched-chain amino acids (BCAAs) are associated with insulin resistance, but it remains unclear whether there is a causal relation between the two. We aimed to disentangle the causal relations by performing a Mendelian randomisation study using genetic variants associated with circulating BCAA levels and insulin resistance as instrumental variables. METHODS We measured circulating BCAA levels in blood plasma by NMR spectroscopy in 1,321 individuals from the ADDITION-PRO cohort. We complemented our analyses by using previously published genome-wide association study (GWAS) results from the Meta-Analyses of Glucose and Insulin-related traits Consortium (MAGIC) (n = 46,186) and from a GWAS of serum BCAA levels (n = 24,925). We used a genetic risk score (GRS), calculated using ten established fasting serum insulin associated variants, as an instrumental variable for insulin resistance. A GRS of three variants increasing circulating BCAA levels was used as an instrumental variable for circulating BCAA levels. RESULTS Fasting plasma BCAA levels were associated with higher HOMA-IR in ADDITION-PRO (β 0.137 [95% CI 0.08, 0.19] p = 6 × 10-7). However, the GRS for circulating BCAA levels was not associated with fasting insulin levels or HOMA-IR in ADDITION-PRO (β -0.011 [95% CI -0.053, 0.032] p = 0.6 and β -0.011 [95% CI -0.054, 0.031] p = 0.6, respectively) or in GWAS results for HOMA-IR from MAGIC (β for valine-increasing GRS -0.012 [95% CI -0.069, 0.045] p = 0.7). By contrast, the insulin-resistance-increasing GRS was significantly associated with increased BCAA levels in ADDITION-PRO (β 0.027 [95% CI 0.005, 0.048] p = 0.01) and in GWAS results for serum BCAA levels (β 1.22 [95% CI 0.71, 1.73] p = 4 × 10-6, β 0.96 [95% CI 0.45, 1.47] p = 3 × 10-4, and β 0.67 [95% CI 0.16, 1.18] p = 0.01 for isoleucine, leucine and valine levels, respectively) and instrumental variable analyses in ADDITION-PRO indicated that HOMA-IR is causally related to higher circulating fasting BCAA levels (β 0.73 [95% CI 0.26, 1.19] p = 0.002). CONCLUSIONS/INTERPRETATION Our results suggest that higher BCAA levels do not have a causal effect on insulin resistance while increased insulin resistance drives higher circulating fasting BCAA levels.
Collapse
Affiliation(s)
- Yuvaraj Mahendran
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark
- The Danish Diabetes Academy, Odense, Denmark
| | - Anna Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark
| | - Christian T Have
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark
| | - Kristine H Allin
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark
| | - Daniel R Witte
- The Danish Diabetes Academy, Odense, Denmark
- Institute of Public Health, University of Aarhus, Aarhus, Denmark
| | | | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, 2100, Copenhagen, Denmark.
| |
Collapse
|
290
|
|
291
|
Ng MCY, Graff M, Lu Y, Justice AE, Mudgal P, Liu CT, Young K, Yanek LR, Feitosa MF, Wojczynski MK, Rand K, Brody JA, Cade BE, Dimitrov L, Duan Q, Guo X, Lange LA, Nalls MA, Okut H, Tajuddin SM, Tayo BO, Vedantam S, Bradfield JP, Chen G, Chen WM, Chesi A, Irvin MR, Padhukasahasram B, Smith JA, Zheng W, Allison MA, Ambrosone CB, Bandera EV, Bartz TM, Berndt SI, Bernstein L, Blot WJ, Bottinger EP, Carpten J, Chanock SJ, Chen YDI, Conti DV, Cooper RS, Fornage M, Freedman BI, Garcia M, Goodman PJ, Hsu YHH, Hu J, Huff CD, Ingles SA, John EM, Kittles R, Klein E, Li J, McKnight B, Nayak U, Nemesure B, Ogunniyi A, Olshan A, Press MF, Rohde R, Rybicki BA, Salako B, Sanderson M, Shao Y, Siscovick DS, Stanford JL, Stevens VL, Stram A, Strom SS, Vaidya D, Witte JS, Yao J, Zhu X, Ziegler RG, Zonderman AB, Adeyemo A, Ambs S, Cushman M, Faul JD, Hakonarson H, Levin AM, Nathanson KL, Ware EB, Weir DR, Zhao W, Zhi D, The Bone Mineral Density in Childhood Study (BMDCS) Group, Arnett DK, Grant SFA, Kardia SLR, Oloapde OI, Rao DC, Rotimi CN, Sale MM, Williams LK, Zemel BS, Becker DM, Borecki IB, et alNg MCY, Graff M, Lu Y, Justice AE, Mudgal P, Liu CT, Young K, Yanek LR, Feitosa MF, Wojczynski MK, Rand K, Brody JA, Cade BE, Dimitrov L, Duan Q, Guo X, Lange LA, Nalls MA, Okut H, Tajuddin SM, Tayo BO, Vedantam S, Bradfield JP, Chen G, Chen WM, Chesi A, Irvin MR, Padhukasahasram B, Smith JA, Zheng W, Allison MA, Ambrosone CB, Bandera EV, Bartz TM, Berndt SI, Bernstein L, Blot WJ, Bottinger EP, Carpten J, Chanock SJ, Chen YDI, Conti DV, Cooper RS, Fornage M, Freedman BI, Garcia M, Goodman PJ, Hsu YHH, Hu J, Huff CD, Ingles SA, John EM, Kittles R, Klein E, Li J, McKnight B, Nayak U, Nemesure B, Ogunniyi A, Olshan A, Press MF, Rohde R, Rybicki BA, Salako B, Sanderson M, Shao Y, Siscovick DS, Stanford JL, Stevens VL, Stram A, Strom SS, Vaidya D, Witte JS, Yao J, Zhu X, Ziegler RG, Zonderman AB, Adeyemo A, Ambs S, Cushman M, Faul JD, Hakonarson H, Levin AM, Nathanson KL, Ware EB, Weir DR, Zhao W, Zhi D, The Bone Mineral Density in Childhood Study (BMDCS) Group, Arnett DK, Grant SFA, Kardia SLR, Oloapde OI, Rao DC, Rotimi CN, Sale MM, Williams LK, Zemel BS, Becker DM, Borecki IB, Evans MK, Harris TB, Hirschhorn JN, Li Y, Patel SR, Psaty BM, Rotter JI, Wilson JG, Bowden DW, Cupples LA, Haiman CA, Loos RJF, North KE. Discovery and fine-mapping of adiposity loci using high density imputation of genome-wide association studies in individuals of African ancestry: African Ancestry Anthropometry Genetics Consortium. PLoS Genet 2017; 13:e1006719. [PMID: 28430825 PMCID: PMC5419579 DOI: 10.1371/journal.pgen.1006719] [Show More Authors] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 05/05/2017] [Accepted: 03/29/2017] [Indexed: 11/20/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified >300 loci associated with measures of adiposity including body mass index (BMI) and waist-to-hip ratio (adjusted for BMI, WHRadjBMI), but few have been identified through screening of the African ancestry genomes. We performed large scale meta-analyses and replications in up to 52,895 individuals for BMI and up to 23,095 individuals for WHRadjBMI from the African Ancestry Anthropometry Genetics Consortium (AAAGC) using 1000 Genomes phase 1 imputed GWAS to improve coverage of both common and low frequency variants in the low linkage disequilibrium African ancestry genomes. In the sex-combined analyses, we identified one novel locus (TCF7L2/HABP2) for WHRadjBMI and eight previously established loci at P < 5×10−8: seven for BMI, and one for WHRadjBMI in African ancestry individuals. An additional novel locus (SPRYD7/DLEU2) was identified for WHRadjBMI when combined with European GWAS. In the sex-stratified analyses, we identified three novel loci for BMI (INTS10/LPL and MLC1 in men, IRX4/IRX2 in women) and four for WHRadjBMI (SSX2IP, CASC8, PDE3B and ZDHHC1/HSD11B2 in women) in individuals of African ancestry or both African and European ancestry. For four of the novel variants, the minor allele frequency was low (<5%). In the trans-ethnic fine mapping of 47 BMI loci and 27 WHRadjBMI loci that were locus-wide significant (P < 0.05 adjusted for effective number of variants per locus) from the African ancestry sex-combined and sex-stratified analyses, 26 BMI loci and 17 WHRadjBMI loci contained ≤ 20 variants in the credible sets that jointly account for 99% posterior probability of driving the associations. The lead variants in 13 of these loci had a high probability of being causal. As compared to our previous HapMap imputed GWAS for BMI and WHRadjBMI including up to 71,412 and 27,350 African ancestry individuals, respectively, our results suggest that 1000 Genomes imputation showed modest improvement in identifying GWAS loci including low frequency variants. Trans-ethnic meta-analyses further improved fine mapping of putative causal variants in loci shared between the African and European ancestry populations. Genome-wide association studies (GWAS) have identified >300 genetic regions that influence body size and shape as measured by body mass index (BMI) and waist-to-hip ratio (WHR), respectively, but few have been identified in populations of African ancestry. We conducted large scale high coverage GWAS and replication of these traits in 52,895 and 23,095 individuals of African ancestry, respectively, followed by additional replication in European populations. We identified 10 genome-wide significant loci in all individuals, and an additional seven loci by analyzing men and women separately. We combined African and European ancestry GWAS and were able to narrow down 43 out of 74 African ancestry associated genetic regions to contain small number of putative causal variants. Our results highlight the improvement of applying high density genome coverage and combining multiple ancestries in the identification and refinement of location of genetic regions associated with adiposity traits.
Collapse
Affiliation(s)
- Maggie C. Y. Ng
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icachn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Poorva Mudgal
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | - Kristin Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Lisa R. Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis MO, United States of America
| | - Mary K. Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis MO, United States of America
| | - Kristin Rand
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Brian E. Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Latchezar Dimitrov
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Qing Duan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Leslie A. Lange
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Michael A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
- Data Tecnica International, Glen Echo, MD, United States of America
| | - Hayrettin Okut
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Salman M. Tajuddin
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| | - Sailaja Vedantam
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | - Jonathan P. Bradfield
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Wei-Min Chen
- Department of Public Health Sciences and Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Alessandra Chesi
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Badri Padhukasahasram
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, United States of America
| | - Jennifer A. Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Matthew A. Allison
- Division of Preventive Medicine, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, United States of America
| | - Christine B. Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - Elisa V. Bandera
- Department of Population Science, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States of America
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Departments of Medicine and Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States of America
| | - Leslie Bernstein
- Beckman Research Institute of the City of Hope, Duarte, CA, United States of America
| | - William J. Blot
- International Epidemiology Institute, Rockville, MD, United States of America
| | - Erwin P. Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icachn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - John Carpten
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States of America
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - David V. Conti
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| | - Myriam Fornage
- Center for Human Genetics, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Barry I. Freedman
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Melissa Garcia
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Phyllis J. Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Yu-Han H. Hsu
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA, United States of America
| | - Jennifer Hu
- Sylvester Comprehensive Cancer Center, University of Miami Leonard Miller School of Medicine, Miami, FL, United States of America
- Department of Public Health Sciences, University of Miami Leonard Miller School of Medicine, Miami, FL, United States of America
| | - Chad D. Huff
- Department of Epidemiology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Sue A. Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States of America
| | - Esther M. John
- Cancer Prevention Institute of California, Fremont, CA, United States of America
- Department of Health Research and Policy (Epidemiology) and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Rick Kittles
- Division of Urology, Department of Surgery, The University of Arizona, Tucson, AZ, United States of America
| | - Eric Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, United States of America
| | - Jin Li
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Barbara McKnight
- Cardiovascular Health Research Unit, Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Uma Nayak
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Barbara Nemesure
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States of America
| | | | - Andrew Olshan
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Michael F. Press
- Department of Pathology and Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States of America
| | - Rebecca Rohde
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Benjamin A. Rybicki
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States of America
| | | | - Maureen Sanderson
- Department of Family and Community Medicine, Meharry Medical College, Nashville, TN, United States of America
| | - Yaming Shao
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - David S. Siscovick
- The New York Academy of Medicine, New York, NY, United States of America
| | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States of America
| | - Victoria L. Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, United States of America
| | - Alex Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Sara S. Strom
- Department of Epidemiology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Dhananjay Vaidya
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - John S. Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States of America
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, United States of America
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Regina G. Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Alan B. Zonderman
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, MD, United States of America
| | - Mary Cushman
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, United States of America
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States of America
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Albert M. Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States of America
| | - Katherine L. Nathanson
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Erin B. Ware
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States of America
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States of America
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Degui Zhi
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | | | - Donna K. Arnett
- School of Public Health, University of Kentucky, Lexington, KY, United States of America
| | - Struan F. A. Grant
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Division of Endocrinology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Sharon L. R. Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Olufunmilayo I. Oloapde
- Center for Clinical Cancer Genetics, Department of Medicine and Human Genetics, University of Chicago, Chicago, IL, United States of America
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Charles N. Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Michele M. Sale
- Department of Public Health Sciences and Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - L. Keoki Williams
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, United States of America
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States of America
| | - Babette S. Zemel
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Diane M. Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis MO, United States of America
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Inc, United States of America
| | - Michele K. Evans
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Tamara B. Harris
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Joel N. Hirschhorn
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Departments of Genetics and Pediatrics, Harvard Medical School, Boston, MA, United States of America
| | - Yun Li
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Sanjay R. Patel
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, United States of America
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
- Division of Genomic Outcomes, Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Los Angeles, CA, United States of America
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Donald W. Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
- NHLBI Framingham Heart Study, Framingham, MA, United States of America
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States of America
- * E-mail: (CAH); (RJFL); (KEN)
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icachn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, NY, United States of America
- * E-mail: (CAH); (RJFL); (KEN)
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
- * E-mail: (CAH); (RJFL); (KEN)
| |
Collapse
|
292
|
Recent progress in genetics, epigenetics and metagenomics unveils the pathophysiology of human obesity. Clin Sci (Lond) 2017; 130:943-86. [PMID: 27154742 DOI: 10.1042/cs20160136] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/24/2016] [Indexed: 12/19/2022]
Abstract
In high-, middle- and low-income countries, the rising prevalence of obesity is the underlying cause of numerous health complications and increased mortality. Being a complex and heritable disorder, obesity results from the interplay between genetic susceptibility, epigenetics, metagenomics and the environment. Attempts at understanding the genetic basis of obesity have identified numerous genes associated with syndromic monogenic, non-syndromic monogenic, oligogenic and polygenic obesity. The genetics of leanness are also considered relevant as it mirrors some of obesity's aetiologies. In this report, we summarize ten genetically elucidated obesity syndromes, some of which are involved in ciliary functioning. We comprehensively review 11 monogenic obesity genes identified to date and their role in energy maintenance as part of the leptin-melanocortin pathway. With the emergence of genome-wide association studies over the last decade, 227 genetic variants involved in different biological pathways (central nervous system, food sensing and digestion, adipocyte differentiation, insulin signalling, lipid metabolism, muscle and liver biology, gut microbiota) have been associated with polygenic obesity. Advances in obligatory and facilitated epigenetic variation, and gene-environment interaction studies have partly accounted for the missing heritability of obesity and provided additional insight into its aetiology. The role of gut microbiota in obesity pathophysiology, as well as the 12 genes associated with lipodystrophies is discussed. Furthermore, in an attempt to improve future studies and merge the gap between research and clinical practice, we provide suggestions on how high-throughput '-omic' data can be integrated in order to get closer to the new age of personalized medicine.
Collapse
|
293
|
Davison JM, Lickwar CR, Song L, Breton G, Crawford GE, Rawls JF. Microbiota regulate intestinal epithelial gene expression by suppressing the transcription factor Hepatocyte nuclear factor 4 alpha. Genome Res 2017; 27:1195-1206. [PMID: 28385711 PMCID: PMC5495071 DOI: 10.1101/gr.220111.116] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
Microbiota influence diverse aspects of intestinal physiology and disease in part by controlling tissue-specific transcription of host genes. However, host genomic mechanisms mediating microbial control of intestinal gene expression are poorly understood. Hepatocyte nuclear factor 4 (HNF4) is the most ancient family of nuclear receptor transcription factors with important roles in human metabolic and inflammatory bowel diseases, but a role in host response to microbes is unknown. Using an unbiased screening strategy, we found that zebrafish Hnf4a specifically binds and activates a microbiota-suppressed intestinal epithelial transcriptional enhancer. Genetic analysis revealed that zebrafish hnf4a activates nearly half of the genes that are suppressed by microbiota, suggesting microbiota negatively regulate Hnf4a. In support, analysis of genomic architecture in mouse intestinal epithelial cells disclosed that microbiota colonization leads to activation or inactivation of hundreds of enhancers along with drastic genome-wide reduction of HNF4A and HNF4G occupancy. Interspecies meta-analysis suggested interactions between HNF4A and microbiota promote gene expression patterns associated with human inflammatory bowel diseases. These results indicate a critical and conserved role for HNF4A in maintaining intestinal homeostasis in response to microbiota.
Collapse
Affiliation(s)
- James M Davison
- Department of Molecular Genetics and Microbiology, Center for the Genomics of Microbial Systems, Duke University, Durham, North Carolina 27710, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Colin R Lickwar
- Department of Molecular Genetics and Microbiology, Center for the Genomics of Microbial Systems, Duke University, Durham, North Carolina 27710, USA
| | - Lingyun Song
- Department of Pediatrics, Division of Medical Genetics, Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, USA
| | - Ghislain Breton
- Department of Integrative Biology and Pharmacology, McGovern Medical School, Houston, Texas 77030, USA
| | - Gregory E Crawford
- Department of Pediatrics, Division of Medical Genetics, Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Center for the Genomics of Microbial Systems, Duke University, Durham, North Carolina 27710, USA
| |
Collapse
|
294
|
Alam I, Reilly AM, Alkhouli M, Gerard-O'Riley RL, Kasipathi C, Oakes DK, Wright WB, Acton D, McQueen AK, Patel B, Lim KE, Robling AG, Econs MJ. Bone Mass and Strength are Significantly Improved in Mice Overexpressing Human WNT16 in Osteocytes. Calcif Tissue Int 2017; 100:361-373. [PMID: 28013361 PMCID: PMC5337173 DOI: 10.1007/s00223-016-0225-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/16/2016] [Indexed: 11/26/2022]
Abstract
Recently, we demonstrated that osteoblast-specific overexpression of human WNT16 increased both cortical and trabecular bone mass and structure in mice. To further identify the cell-specific role of Wnt16 in bone homeostasis, we created transgenic (TG) mice overexpressing human WNT16 in osteocytes using Dmp1 promoter (Dmp1-hWNT16 TG) on C57BL/6 (B6) background. We analyzed bone phenotypes and serum bone biomarkers, performed gene expression analysis and measured dynamic bone histomorphometry in Dmp1-hWNT16 TG and wild-type (WT) mice. Compared to WT mice, Dmp1-hWNT16 TG mice exhibited significantly higher whole-body, spine and femoral aBMD, BMC and trabecular (BV/TV, Tb.N, and Tb.Th) and cortical (bone area and thickness) parameters in both male and female at 12 weeks of age. Femur stiffness and ultimate force were also significantly improved in the Dmp1-hWNT16 TG female mice, compared to sex-matched WT littermates. In addition, female Dmp1-hWNT16 TG mice displayed significantly higher MS/BS, MAR and BFR/BS compared to the WT mice. Gene expression analysis demonstrated significantly higher mRNA level of Alp in both male and female Dmp1-hWNT16 TG mice and significantly higher levels of Osteocalcin, Opg and Rankl in the male Dmp1-hWNT16 TG mice in bone tissue compared to sex-matched WT mice. These results indicate that WNT16 plays a critical role for acquisition of both cortical and trabecular bone mass and strength. Strategies designed to use WNT16 as a target for therapeutic interventions will be valuable to treat osteoporosis and other low bone mass conditions.
Collapse
Affiliation(s)
- Imranul Alam
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA.
| | - Austin M Reilly
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Mohammed Alkhouli
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Rita L Gerard-O'Riley
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Charishma Kasipathi
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Dana K Oakes
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Weston B Wright
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Dena Acton
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Amie K McQueen
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Bhavmik Patel
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
| | - Kyung-Eun Lim
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael J Econs
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 West Michigan St, CL459, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
295
|
Wang Y, Wactawski-Wende J, Sucheston-Campbell LE, Preus L, Hovey KM, Nie J, Jackson RD, Handelman SK, Nassir R, Crandall CJ, Ochs-Balcom HM. The influence of genetic susceptibility and calcium plus vitamin D supplementation on fracture risk. Am J Clin Nutr 2017; 105:970-979. [PMID: 28148500 PMCID: PMC5366049 DOI: 10.3945/ajcn.116.144550] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/04/2017] [Indexed: 01/21/2023] Open
Abstract
Background: Fracture is a complex trait, affected by both genetic and environmental factors. A meta-analysis of genome-wide association studies (GWASs) identified multiple bone mineral density (BMD) and fracture-associated loci.Objective: We conducted a study to evaluate whether fracture genetic risk score (Fx-GRS) and bone mineral density genetic risk score (BMD-GRS) modify the association between the intake of calcium with vitamin D (CaD) and fracture risk.Design: Data from 5823 white postmenopausal women from the Women's Health Initiative CaD randomized trial were included. Participants received 1000 mg elemental Ca with 400 IU vitamin D3/d or placebo (median follow-up: 6.5 y). Total fracture was defined as first fracture of any type. We computed the Fx-GRS with 16 fracture- and BMD-associated variants, and the BMD-GRS with 50 BMD-associated variants. We used Cox regression and a case-only approach to test for multiplicative interaction. Additive interaction was assessed with the relative excess risk due to interaction (RERI). We analyzed genetic risk score as a continuous variable and a categorical variable based on quartile (quartile 1, quartiles 2-3, and quartile 4).Results: We observed no interaction between the Fx-GRS and CaD on fracture risk; however, we observed a significant multiplicative interaction between the BMD-GRS and CaD assignment (P-interaction = 0.01). In addition, there was a significant negative additive interaction between placebo assignment and higher BMD-GRS: quartiles 2-3, PRERI = 0.03; quartile 4, PRERI = 0.03. In a stratified analysis, the protective effect of CaD on fracture risk was observed in women in the lowest BMD-GRS quartile (HR: 0.60, 95% CI: 0.44, 0.81) but not in women with a higher BMD-GRS.Conclusions: We observed significant effects of CaD intake on fracture risk only in women with the lowest genetic predisposition to low BMD. Future large-scale studies with functional characterization of GWAS findings are warranted to assess the utility of genetic risk score in analysis of risks and benefits of CaD for bone.
Collapse
Affiliation(s)
- Youjin Wang
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY
| | | | - Leah Preus
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY
- Department of Cancer Prevention and Control, Division of Cancer Prevention and Population Sciences, Roswell Park Cancer Institute, Buffalo, NY
| | - Kathleen M Hovey
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY
| | - Jing Nie
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY
| | - Rebecca D Jackson
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, and
| | - Samuel K Handelman
- Center for Pharmacogenomics, Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH
| | - Rami Nassir
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA; and
| | - Carolyn J Crandall
- Division of General Internal Medicine and Health Sciences Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Heather M Ochs-Balcom
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY;
| |
Collapse
|
296
|
Maridas DE, DeMambro VE, Le PT, Nagano K, Baron R, Mohan S, Rosen CJ. IGFBP-4 regulates adult skeletal growth in a sex-specific manner. J Endocrinol 2017; 233:131-144. [PMID: 28184001 PMCID: PMC5425953 DOI: 10.1530/joe-16-0673] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 02/09/2017] [Indexed: 01/19/2023]
Abstract
Insulin-like growth factor-1 (IGF-1) and its binding proteins are critical mediators of skeletal growth. Insulin-like growth factor-binding protein 4 (IGFBP-4) is highly expressed in osteoblasts and inhibits IGF-1 actions in vitro Yet, in vivo studies suggest that it could potentiate IGF-1 and IGF-2 actions. In this study, we hypothesized that IGFBP-4 might potentiate the actions of IGF-1 on the skeleton. To test this, we comprehensively studied 8- and 16-week-old Igfbp4-/- mice. Both male and female adult Igfbp4-/- mice had marked growth retardation with reductions in body weight, body and femur lengths, fat proportion and lean mass at 8 and 16 weeks. Marked reductions in aBMD and aBMC were observed in 16-week-old Igfbp4-/- females, but not in males. Femoral trabecular BV/TV and thickness, cortical fraction and thickness in 16-week-old Igfbp4-/- females were significantly reduced. However, surprisingly, males had significantly more trabeculae with higher connectivity density than controls. Concordantly, histomorphometry revealed higher bone resorption and lower bone formation in Igfbp4-/- females. In contrast, Igfbp4-/- males had lower mineralized surface/bone surface. Femoral expression of Sost and circulating levels of sclerostin were reduced but only in Igfbp4-/- males. Bone marrow stromal cultures from mutants showed increased osteogenesis, whereas osteoclastogenesis was markedly increased in cells from Igfbp4-/- females but decreased in males. In sum, our results indicate that loss of Igfbp4 affects mesenchymal stromal cell differentiation, regulates osteoclastogenesis and influences both skeletal development and adult bone maintenance. Thus, IGFBP-4 modulates the skeleton in a gender-specific manner, acting as both a cell autonomous and cell non-autonomous factor.
Collapse
Affiliation(s)
- David E Maridas
- Maine Medical Center Research InstituteScarborough, Maine, USA
| | | | - Phuong T Le
- Maine Medical Center Research InstituteScarborough, Maine, USA
| | - Kenichi Nagano
- Harvard School of Dental MedicineBoston, Massachusetts, USA
| | - Roland Baron
- Harvard School of Dental MedicineBoston, Massachusetts, USA
| | | | | |
Collapse
|
297
|
Dessauer CW, Watts VJ, Ostrom RS, Conti M, Dove S, Seifert R. International Union of Basic and Clinical Pharmacology. CI. Structures and Small Molecule Modulators of Mammalian Adenylyl Cyclases. Pharmacol Rev 2017; 69:93-139. [PMID: 28255005 PMCID: PMC5394921 DOI: 10.1124/pr.116.013078] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adenylyl cyclases (ACs) generate the second messenger cAMP from ATP. Mammalian cells express nine transmembrane AC (mAC) isoforms (AC1-9) and a soluble AC (sAC, also referred to as AC10). This review will largely focus on mACs. mACs are activated by the G-protein Gαs and regulated by multiple mechanisms. mACs are differentially expressed in tissues and regulate numerous and diverse cell functions. mACs localize in distinct membrane compartments and form signaling complexes. sAC is activated by bicarbonate with physiologic roles first described in testis. Crystal structures of the catalytic core of a hybrid mAC and sAC are available. These structures provide detailed insights into the catalytic mechanism and constitute the basis for the development of isoform-selective activators and inhibitors. Although potent competitive and noncompetitive mAC inhibitors are available, it is challenging to obtain compounds with high isoform selectivity due to the conservation of the catalytic core. Accordingly, caution must be exerted with the interpretation of intact-cell studies. The development of isoform-selective activators, the plant diterpene forskolin being the starting compound, has been equally challenging. There is no known endogenous ligand for the forskolin binding site. Recently, development of selective sAC inhibitors was reported. An emerging field is the association of AC gene polymorphisms with human diseases. For example, mutations in the AC5 gene (ADCY5) cause hyperkinetic extrapyramidal motor disorders. Overall, in contrast to the guanylyl cyclase field, our understanding of the (patho)physiology of AC isoforms and the development of clinically useful drugs targeting ACs is still in its infancy.
Collapse
Affiliation(s)
- Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Val J Watts
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Rennolds S Ostrom
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Marco Conti
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Stefan Dove
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Roland Seifert
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| |
Collapse
|
298
|
GeneImp: Fast Imputation to Large Reference Panels Using Genotype Likelihoods from Ultralow Coverage Sequencing. Genetics 2017; 206:91-104. [PMID: 28348060 DOI: 10.1534/genetics.117.200063] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/20/2017] [Indexed: 01/14/2023] Open
Abstract
We address the task of genotype imputation to a dense reference panel given genotype likelihoods computed from ultralow coverage sequencing as inputs. In this setting, the data have a high-level of missingness or uncertainty, and are thus more amenable to a probabilistic representation. Most existing imputation algorithms are not well suited for this situation, as they rely on prephasing for computational efficiency, and, without definite genotype calls, the prephasing task becomes computationally expensive. We describe GeneImp, a program for genotype imputation that does not require prephasing and is computationally tractable for whole-genome imputation. GeneImp does not explicitly model recombination, instead it capitalizes on the existence of large reference panels-comprising thousands of reference haplotypes-and assumes that the reference haplotypes can adequately represent the target haplotypes over short regions unaltered. We validate GeneImp based on data from ultralow coverage sequencing (0.5×), and compare its performance to the most recent version of BEAGLE that can perform this task. We show that GeneImp achieves imputation quality very close to that of BEAGLE, using one to two orders of magnitude less time, without an increase in memory complexity. Therefore, GeneImp is the first practical choice for whole-genome imputation to a dense reference panel when prephasing cannot be applied, for instance, in datasets produced via ultralow coverage sequencing. A related future application for GeneImp is whole-genome imputation based on the off-target reads from deep whole-exome sequencing.
Collapse
|
299
|
Exploring the relationship between α-actinin-3 deficiency and obesity in mice and humans. Int J Obes (Lond) 2017; 41:1154-1157. [PMID: 28293018 PMCID: PMC5504447 DOI: 10.1038/ijo.2017.72] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/06/2017] [Accepted: 02/26/2017] [Indexed: 11/08/2022]
Abstract
Obesity is a worldwide health crisis, and the identification of genetic modifiers of weight gain is crucial in understanding this complex disorder. A common null polymorphism in the fast fiber-specific gene ACTN3 (R577X) is known to influence skeletal muscle function and metabolism. α-Actinin-3 deficiency occurs in an estimated 1.5 billion people worldwide, and results in reduced muscle strength and a shift towards a more efficient oxidative metabolism. The X-allele has undergone strong positive selection during recent human evolution, and in this study, we sought to determine whether ACTN3 genotype influences weight gain and obesity in mice and humans. An Actn3 KO mouse has been generated on two genetic backgrounds (129X1/SvJ and C57BL/6J) and fed a high-fat diet (HFD, 45% calories from fat). Anthropomorphic features (including body weight) were examined and show that Actn3 KO 129X1/SvJ mice gained less weight compared to WT. In addition, six independent human cohorts were genotyped for ACTN3 R577X (Rs1815739) and body mass index (BMI), waist-to-hip ratio-adjusted BMI (WHRadjBMI) and obesity-related traits were assessed. In humans, ACTN3 genotype alone does not contribute to alterations in BMI or obesity.
Collapse
|
300
|
Abstract
AbstractBody mass and fat intake are multifactorial traits that have genetic and environmental components. The gene with the greatest effect on body mass is FTO (fat mass and obesity-associated), but several studies have shown that the effect of FTO (and of other genes) on body mass can be modified by the intake of nutrients. The so-called gene–environment interactions may also be important for the effectiveness of weight-loss strategies. Food choices, and thus fat intake, depend to some extent on individual preferences. The most important biological component of food preference is taste, and the role of fat sensitivity in fat intake has recently been pointed out. Relatively few studies have analysed the genetic components of fat intake or fatty acid sensitivity in terms of their relation to obesity. It has been proposed that decreased oral fatty acid sensitivity leads to increased fat intake and thus increased body mass. One of the genes that affect fatty acid sensitivity is CD36 (cluster of differentiation 36). However, little is known so far about the genetic component of fat sensing. We performed a literature review to identify the state of knowledge regarding the genetics of fat intake and its relation to body-mass determination, and to identify the priorities for further investigations.
Collapse
|