951
|
Huang Z, Lin H, Fellay J, Kutalik Z, Hubaux JP. SQC: secure quality control for meta-analysis of genome-wide association studies. Bioinformatics 2017; 33:2273-2280. [DOI: 10.1093/bioinformatics/btx193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 03/31/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Zhicong Huang
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Huang Lin
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jacques Fellay
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute of Social and Preventive Medicine, University Hospital Lausanne (CHUV), Lausanne, Switzerland
| | - Jean-Pierre Hubaux
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
952
|
Otto JM, Gizer IR, Ellingson JM, Wilhelmsen KC. Genetic variation in the exome: Associations with alcohol and tobacco co-use. PSYCHOLOGY OF ADDICTIVE BEHAVIORS 2017; 31:354-366. [PMID: 28368157 DOI: 10.1037/adb0000270] [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] [Indexed: 12/11/2022]
Abstract
Shared genetic factors represent one underlying mechanism thought to contribute to high rates of alcohol and tobacco co-use and dependence. Common variants identified by molecular genetic studies tend to confer only small disease risk, and rare protein-coding variants are posited to contribute to disease risk, as well. However, given that genotyping technologies allowing for their inclusion in association studies have only recently become available, the magnitude of their contribution is poorly understood. The current study examined genetic variation in protein-coding regions (i.e., the exome) for associations with measures of lifetime alcohol and tobacco co-use. Participants from the UCSF Family Alcoholism Study (N = 1,862) were genotyped using an exome-focused genotyping array, and assessed for DSM-IV diagnoses of alcohol and tobacco dependence and quantitative consumption measures using a modified version of the Semi-Structured Assessment for the Genetics of Alcoholism. Analyses included single variant, gene-based, and pathway-based tests of association. One EMR3 variant and a pathway related to genes upregulated in mesenchymal stem cells during the late phase of adipogenesis met criteria for statistical significance. Suggestive associations were consistent with previous findings from studies of substance use and dependence, including variants in the CHRNA5-CHRNA3-CHRNB4 gene cluster with cigarettes smoked per day. Further, several variants and genes demonstrated suggestive association across phenotypes, suggesting that shared genetic factors may underlie risk for increased levels of alcohol and tobacco use, as well as psychopathology more broadly, providing insight into our understanding of the genetic architecture underlying these traits. (PsycINFO Database Record
Collapse
Affiliation(s)
- Jacqueline M Otto
- Department of Psychological Sciences, University of Missouri-Columbia
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri-Columbia
| | | | - Kirk C Wilhelmsen
- Department of Genetics and Neurology, University of North Carolina at Chapel Hill
| |
Collapse
|
953
|
Automated Phenotyping Indicates Pupal Size in Drosophila Is a Highly Heritable Trait with an Apparent Polygenic Basis. G3-GENES GENOMES GENETICS 2017; 7:1277-1286. [PMID: 28258111 PMCID: PMC5386876 DOI: 10.1534/g3.117.039883] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The intense focus on studying human height has done more than any other genetic analysis to advance our understanding of the heritability of highly complex phenotypes. Here, we describe in detail the properties of a previously unexplored trait in Drosophila melanogaster that shares many salient properties with human height. The total length of the pupal case varies between 2.8 and 3.9 mm among natural variants, and we report that it is among the most heritable traits reported in this species. We have developed a simple semiautomatic phenotyping system with which a single operator can reliably score >5000 individuals in a day. The precision of the automated system is 0.042 mm (± 0.030 SD). All phenotyped individuals are available to be mated in subsequent generations or uniquely archived for future molecular work. We report both broad sense and narrow sense heritability estimates for two biologically distinct data sets. Narrow sense heritability (h2) ranged from 0.44 to 0.50, and broad sense heritability (H2) ranged from 0.58 to 0.61. We present results for mapping the trait in 195 recombinant inbred lines, which suggests that there are no loci with >10% effect size in this panel. We propose that pupal size genetics in Drosophila could represent a model complex trait amenable to deep genetic dissection using the automated system described.
Collapse
|
954
|
Time-Variant Genetic Effects as a Cause for Preterm Birth: Insights from a Population of Maternal Cousins in Sweden. G3-GENES GENOMES GENETICS 2017; 7:1349-1356. [PMID: 28250013 PMCID: PMC5386882 DOI: 10.1534/g3.116.038612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Preterm delivery (PTD) is the leading cause of neonatal mortality worldwide, yet its etiology remains largely unexplained. We propose that the genetic factors controlling this trait could act in a nonuniform manner during pregnancy, with each factor having a unique “window of sensitivity.” We test this hypothesis by modeling the distribution of gestational ages (GAs) observed in maternal cousins from the Swedish Medical Birth Register (MBR) (n = 35,541 pairs). The models were built using a time-to-event framework, with simulated genetic factors that increase the hazard of birth either uniformly across the pregnancy (constant effect) or only in particular windows (varying effect). By including various combinations of these factors, we obtained four models that were then optimized and compared. Best fit to the clinical data was observed when most of the factors had time-variant effects, independently of the number of loci simulated. Finally, power simulations were performed to assess the ability to discover varying-effect loci by usual methods for genome-wide association testing. We believe that the tools and concepts presented here should prove useful for the design of future studies of PTD and provide new insights into the genetic architecture determining human GA.
Collapse
|
955
|
Gage SH, Jones HJ, Burgess S, Bowden J, Davey Smith G, Zammit S, Munafò MR. Assessing causality in associations between cannabis use and schizophrenia risk: a two-sample Mendelian randomization study. Psychol Med 2017; 47:971-980. [PMID: 27928975 PMCID: PMC5341491 DOI: 10.1017/s0033291716003172] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Observational associations between cannabis and schizophrenia are well documented, but ascertaining causation is more challenging. We used Mendelian randomization (MR), utilizing publicly available data as a method for ascertaining causation from observational data. METHOD We performed bi-directional two-sample MR using summary-level genome-wide data from the International Cannabis Consortium (ICC) and the Psychiatric Genomics Consortium (PGC2). Single nucleotide polymorphisms (SNPs) associated with cannabis initiation (p < 10-5) and schizophrenia (p < 5 × 10-8) were combined using an inverse-variance-weighted fixed-effects approach. We also used height and education genome-wide association study data, representing negative and positive control analyses. RESULTS There was some evidence consistent with a causal effect of cannabis initiation on risk of schizophrenia [odds ratio (OR) 1.04 per doubling odds of cannabis initiation, 95% confidence interval (CI) 1.01-1.07, p = 0.019]. There was strong evidence consistent with a causal effect of schizophrenia risk on likelihood of cannabis initiation (OR 1.10 per doubling of the odds of schizophrenia, 95% CI 1.05-1.14, p = 2.64 × 10-5). Findings were as predicted for the negative control (height: OR 1.00, 95% CI 0.99-1.01, p = 0.90) but weaker than predicted for the positive control (years in education: OR 0.99, 95% CI 0.97-1.00, p = 0.066) analyses. CONCLUSIONS Our results provide some that cannabis initiation increases the risk of schizophrenia, although the size of the causal estimate is small. We find stronger evidence that schizophrenia risk predicts cannabis initiation, possibly as genetic instruments for schizophrenia are stronger than for cannabis initiation.
Collapse
Affiliation(s)
- S. H. Gage
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Experimental Psychology, University of Bristol, Bristol, UK
| | - H. J. Jones
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - S. Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - J. Bowden
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - G. Davey Smith
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - S. Zammit
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - M. R. Munafò
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Experimental Psychology, University of Bristol, Bristol, UK
| |
Collapse
|
956
|
Fang L, Sahana G, Su G, Yu Y, Zhang S, Lund MS, Sørensen P. Integrating Sequence-based GWAS and RNA-Seq Provides Novel Insights into the Genetic Basis of Mastitis and Milk Production in Dairy Cattle. Sci Rep 2017; 7:45560. [PMID: 28358110 PMCID: PMC5372096 DOI: 10.1038/srep45560] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/28/2017] [Indexed: 02/06/2023] Open
Abstract
Connecting genome-wide association study (GWAS) to biological mechanisms underlying complex traits is a major challenge. Mastitis resistance and milk production are complex traits of economic importance in the dairy sector and are associated with intra-mammary infection (IMI). Here, we integrated IMI-relevant RNA-Seq data from Holstein cattle and sequence-based GWAS data from three dairy cattle breeds (i.e., Holstein, Nordic red cattle, and Jersey) to explore the genetic basis of mastitis resistance and milk production using post-GWAS analyses and a genomic feature linear mixed model. At 24 h post-IMI, genes responsive to IMI in the mammary gland were preferentially enriched for genetic variants associated with mastitis resistance rather than milk production. Response genes in the liver were mainly enriched for variants associated with mastitis resistance at an early time point (3 h) post-IMI, whereas responsive genes at later stages were enriched for associated variants with milk production. The up- and down-regulated genes were enriched for associated variants with mastitis resistance and milk production, respectively. The patterns were consistent across breeds, indicating that different breeds shared similarities in the genetic basis of these traits. Our approaches provide a framework for integrating multiple layers of data to understand the genetic architecture underlying complex traits.
Collapse
Affiliation(s)
- Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture &National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China
| | - Goutam Sahana
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Guosheng Su
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Ying Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture &National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China
| | - Shengli Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture &National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China
| | - Mogens Sandø Lund
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Peter Sørensen
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| |
Collapse
|
957
|
Heck A, Milnik A, Vukojevic V, Petrovska J, Egli T, Singer J, Escobar P, Sengstag T, Coynel D, Freytag V, Fastenrath M, Demougin P, Loos E, Hartmann F, Schicktanz N, Delarue Bizzini B, Vogler C, Kolassa IT, Wilker S, Elbert T, Schwede T, Beisel C, Beerenwinkel N, de Quervain DJF, Papassotiropoulos A. Exome sequencing of healthy phenotypic extremes links TROVE2 to emotional memory and PTSD. Nat Hum Behav 2017. [DOI: 10.1038/s41562-017-0081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
958
|
McLaughlin RL, Schijven D, van Rheenen W, van Eijk KR, O'Brien M, Kahn RS, Ophoff RA, Goris A, Bradley DG, Al-Chalabi A, van den Berg LH, Luykx JJ, Hardiman O, Veldink JH. Genetic correlation between amyotrophic lateral sclerosis and schizophrenia. Nat Commun 2017; 8:14774. [PMID: 28322246 PMCID: PMC5364411 DOI: 10.1038/ncomms14774] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 02/03/2017] [Indexed: 12/11/2022] Open
Abstract
We have previously shown higher-than-expected rates of schizophrenia in relatives of patients with amyotrophic lateral sclerosis (ALS), suggesting an aetiological relationship between the diseases. Here, we investigate the genetic relationship between ALS and schizophrenia using genome-wide association study data from over 100,000 unique individuals. Using linkage disequilibrium score regression, we estimate the genetic correlation between ALS and schizophrenia to be 14.3% (7.05-21.6; P=1 × 10-4) with schizophrenia polygenic risk scores explaining up to 0.12% of the variance in ALS (P=8.4 × 10-7). A modest increase in comorbidity of ALS and schizophrenia is expected given these findings (odds ratio 1.08-1.26) but this would require very large studies to observe epidemiologically. We identify five potential novel ALS-associated loci using conditional false discovery rate analysis. It is likely that shared neurobiological mechanisms between these two disorders will engender novel hypotheses in future preclinical and clinical studies.
Collapse
Affiliation(s)
- Russell L. McLaughlin
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin DO2 DK07, Republic of Ireland
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 DK07, Republic of Ireland
| | - Dick Schijven
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Wouter van Rheenen
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Kristel R. van Eijk
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Margaret O'Brien
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin DO2 DK07, Republic of Ireland
| | - René S. Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Roel A. Ophoff
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California 90095, USA
| | - An Goris
- Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven—University of Leuven, Leuven B-3000, Belgium
| | - Daniel G. Bradley
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 DK07, Republic of Ireland
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London WC2R 2LS, UK
| | - Leonard H. van den Berg
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Jurjen J. Luykx
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
- Department of Psychiatry, Hospital Network Antwerp (ZNA) Stuivenberg and Sint Erasmus, Antwerp 2020, Belgium
| | - Orla Hardiman
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 DK07, Republic of Ireland
| | - Jan H. Veldink
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| |
Collapse
|
959
|
Porter HF, O’Reilly PF. Multivariate simulation framework reveals performance of multi-trait GWAS methods. Sci Rep 2017; 7:38837. [PMID: 28287610 PMCID: PMC5347376 DOI: 10.1038/srep38837] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/19/2016] [Indexed: 01/22/2023] Open
Abstract
Burgeoning availability of genome-wide association study (GWAS) results and national biobank data has led to growing interest in performing multi-trait genetic analyses. Numerous multi-trait GWAS methods that exploit either summary statistics or individual-level data have been developed, but their relative performance is unclear. Here we develop a simulation framework to model the complex networks underlying multivariate genetic epidemiology, enabling the vast model space of genetic effects on multiple correlated traits to be explored systematically. We perform a comprehensive comparison of the leading multi-trait GWAS methods, finding: (1) method performance is highly sensitive to the specific combination of genetic effects and phenotypic correlations, (2) most of the current multivariate methods have remarkably similar statistical power, and (3) multivariate methods may offer a substantial increase in the discovery of genetic variants over the standard univariate approach. We believe our findings offer the clearest picture to date of the relative performance of multi-trait GWAS methods and act as a guide for method selection. We provide a web application and open-source software program implementing our simulation framework, for: (i) further benchmarking of multivariate GWAS methods, (ii) power calculations for multivariate genetic studies, and (iii) generating data for testing any multivariate method in genetic epidemiology.
Collapse
Affiliation(s)
- Heather F. Porter
- MRC SGDP Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, SE5 8AF, UK
| | - Paul F. O’Reilly
- MRC SGDP Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, SE5 8AF, UK
| |
Collapse
|
960
|
Abstract
Parameters of bone geometry such as width, length, and cross-sectional area are major determinants of bone strength. Although these traits are highly heritable, few genes influencing bone geometry have been identified. Here, we dissect a major quantitative trait locus (QTL) influencing femur size. This QTL was originally identified in an F2 cross between the C57BL/6J-hg/hg (HG) and CAST/EiJ strains and was referred to as femur length in high growth mice 2 (Feml2). Feml2 was located on chromosome (Chr.) 9 at ∼20 cM. Here, we show that the HG.CAST-(D9Mit249-D9Mit133)/Ucd congenic strain captures Feml2. In an F2 congenic cross, we fine-mapped the location of Feml2 to an ∼6 Mbp region extending from 57.3 to 63.3 Mbp on Chr. 9. We have identified candidates by mining the complete genome sequence of CAST/EiJ and through allele-specific expression (ASE) analysis of growth plates in C57BL/6J × CAST/EiJ F1 hybrids. Interestingly, we also find that the refined location of Feml2 overlaps a cluster of six independent genome-wide associations for human height. This work provides the foundation for the identification of novel genes affecting bone geometry.
Collapse
|
961
|
Wen X, Pique-Regi R, Luca F. Integrating molecular QTL data into genome-wide genetic association analysis: Probabilistic assessment of enrichment and colocalization. PLoS Genet 2017; 13:e1006646. [PMID: 28278150 PMCID: PMC5363995 DOI: 10.1371/journal.pgen.1006646] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 03/23/2017] [Accepted: 02/21/2017] [Indexed: 01/25/2023] Open
Abstract
We propose a novel statistical framework for integrating the result from molecular quantitative trait loci (QTL) mapping into genome-wide genetic association analysis of complex traits, with the primary objectives of quantitatively assessing the enrichment of the molecular QTLs in complex trait-associated genetic variants and the colocalizations of the two types of association signals. We introduce a natural Bayesian hierarchical model that treats the latent association status of molecular QTLs as SNP-level annotations for candidate SNPs of complex traits. We detail a computational procedure to seamlessly perform enrichment, fine-mapping and colocalization analyses, which is a distinct feature compared to the existing colocalization analysis procedures in the literature. The proposed approach is computationally efficient and requires only summary-level statistics. We evaluate and demonstrate the proposed computational approach through extensive simulation studies and analyses of blood lipid data and the whole blood eQTL data from the GTEx project. In addition, a useful utility from our proposed method enables the computation of expected colocalization signals using simple characteristics of the association data. Using this utility, we further illustrate the importance of enrichment analysis on the ability to discover colocalized signals and the potential limitations of currently available molecular QTL data. The software pipeline that implements the proposed computation procedures, enloc, is freely available at https://github.com/xqwen/integrative. Genome-wide association studies (GWAS) have been tremendously successful in identifying genetic variants that impact complex diseases. However, the roles of such studies in disease etiology remain poorly understood, primarily because a large proportion of the GWAS findings are located in the non-coding region of the genome. Recent advancements in high-throughput sequencing technology enable the systematic investigation of molecular quantitative trait loci (QTLs), which are genetic variants that directly affect molecular phenotypes (e.g., gene expression, transcription factor binding and DNA methylation). Linking molecular QTLs to GWAS findings intuitively represents an important step for interpreting the biological and clinical relevance of the GWAS results. In this paper, we describe a rigorous and efficient computational approach that assesses the enrichment and overlap between the GWAS findings and molecular QTLs. Importantly, we illustrate that the accurate quantification of overlapping between molecular QTL and GWAS signals requires reliable enrichment estimation. Our proposed approach fully accounts for the intrinsic uncertainty embedded in the association analyses of GWAS and molecular QTL mapping, and it outperforms the existing state-of-the-art approaches. Applying the proposed approach to the GWAS data of blood lipid traits and the whole blood expression QTLs (eQTLs) yields some novel biological insights and also illustrates the potential limitations of the currently available molecular QTL data.
Collapse
Affiliation(s)
- Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| |
Collapse
|
962
|
Mancuso N, Shi H, Goddard P, Kichaev G, Gusev A, Pasaniuc B. Integrating Gene Expression with Summary Association Statistics to Identify Genes Associated with 30 Complex Traits. Am J Hum Genet 2017; 100:473-487. [PMID: 28238358 PMCID: PMC5339290 DOI: 10.1016/j.ajhg.2017.01.031] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/23/2017] [Indexed: 01/24/2023] Open
Abstract
Although genome-wide association studies (GWASs) have identified thousands of risk loci for many complex traits and diseases, the causal variants and genes at these loci remain largely unknown. Here, we introduce a method for estimating the local genetic correlation between gene expression and a complex trait and utilize it to estimate the genetic correlation due to predicted expression between pairs of traits. We integrated gene expression measurements from 45 expression panels with summary GWAS data to perform 30 multi-tissue transcriptome-wide association studies (TWASs). We identified 1,196 genes whose expression is associated with these traits; of these, 168 reside more than 0.5 Mb away from any previously reported GWAS significant variant. We then used our approach to find 43 pairs of traits with significant genetic correlation at the level of predicted expression; of these, eight were not found through genetic correlation at the SNP level. Finally, we used bi-directional regression to find evidence that BMI causally influences triglyceride levels and that triglyceride levels causally influence low-density lipoprotein. Together, our results provide insight into the role of gene expression in the susceptibility of complex traits and diseases.
Collapse
Affiliation(s)
- Nicholas Mancuso
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA.
| | - Huwenbo Shi
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Pagé Goddard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Gleb Kichaev
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Alexander Gusev
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bogdan Pasaniuc
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90024, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA.
| |
Collapse
|
963
|
Racimo F, Gokhman D, Fumagalli M, Ko A, Hansen T, Moltke I, Albrechtsen A, Carmel L, Huerta-Sánchez E, Nielsen R. Archaic Adaptive Introgression in TBX15/WARS2. Mol Biol Evol 2017; 34:509-524. [PMID: 28007980 PMCID: PMC5430617 DOI: 10.1093/molbev/msw283] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A recent study conducted the first genome-wide scan for selection in Inuit from Greenland using single nucleotide polymorphism chip data. Here, we report that selection in the region with the second most extreme signal of positive selection in Greenlandic Inuit favored a deeply divergent haplotype that is closely related to the sequence in the Denisovan genome, and was likely introgressed from an archaic population. The region contains two genes, WARS2 and TBX15, and has previously been associated with adipose tissue differentiation and body-fat distribution in humans. We show that the adaptively introgressed allele has been under selection in a much larger geographic region than just Greenland. Furthermore, it is associated with changes in expression of WARS2 and TBX15 in multiple tissues including the adrenal gland and subcutaneous adipose tissue, and with regional DNA methylation changes in TBX15.
Collapse
Affiliation(s)
- Fernando Racimo
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA
| | - David Gokhman
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, Faculty of Science, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem, Israel
| | - Matteo Fumagalli
- Department of Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Amy Ko
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Moltke
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Anders Albrechtsen
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Liran Carmel
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, Faculty of Science, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem, Israel
| | | | - Rasmus Nielsen
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA
- Department of Statistics, University of California Berkeley, Berkeley, CA
- Museum of Natural History, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
964
|
Sabik OL, Farber CR. Using GWAS to identify novel therapeutic targets for osteoporosis. Transl Res 2017; 181:15-26. [PMID: 27837649 PMCID: PMC5357198 DOI: 10.1016/j.trsl.2016.10.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022]
Abstract
Osteoporosis is a common, increasingly prevalent, global health burden characterized by low bone mineral density (BMD) and increased risk of fracture. Despite its significant impact on human health, there is currently a lack of highly effective treatments free of side effects for osteoporosis. Therefore, a major goal in the field is to identify new drug targets. Genetic discovery has been shown to be effective in the unbiased identification of novel drug targets and genome-wide association studies (GWASs) have begun to provide insight into genetic basis of osteoporosis. Over the last decade, GWASs have led to the identification of ∼100 loci associated with BMD and other bone traits related to risk of fracture. However, there have been limited efforts to identify the causal genes underlying the GWAS loci or the mechanisms by which GWAS loci alter bone physiology. In this review, we summarize the current state of the field and discuss strategies for causal gene discovery and the evidence that the novel genes underlying GWAS loci are likely to be a new source of drug targets.
Collapse
Affiliation(s)
- Olivia L Sabik
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Va; Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, Va
| | - Charles R Farber
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Va; Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, Va; Department of Public Health Science, School of Medicine, University of Virginia, Charlottesville, Va.
| |
Collapse
|
965
|
Hobbs BD, de Jong K, Lamontagne M, Bossé Y, Shrine N, Artigas MS, Wain LV, Hall IP, Jackson VE, Wyss AB, London SJ, North KE, Franceschini N, Strachan DP, Beaty TH, Hokanson JE, Crapo JD, Castaldi PJ, Chase RP, Bartz TM, Heckbert SR, Psaty BM, Gharib SA, Zanen P, Lammers JW, Oudkerk M, Groen HJ, Locantore N, Tal-Singer R, Rennard SI, Vestbo J, Timens W, Paré PD, Latourelle JC, Dupuis J, O’Connor GT, Wilk JB, Kim WJ, Lee MK, Oh YM, Vonk JM, de Koning HJ, Leng S, Belinsky SA, Tesfaigzi Y, Manichaikul A, Wang XQ, Rich SS, Barr RG, Sparrow D, Litonjua AA, Bakke P, Gulsvik A, Lahousse L, Brusselle GG, Stricker BH, Uitterlinden AG, Ampleford EJ, Bleecker ER, Woodruff PG, Meyers DA, Qiao D, Lomas DA, Yim JJ, Kim DK, Hawrylkiewicz I, Sliwinski P, Hardin M, Fingerlin TE, Schwartz DA, Postma DS, MacNee W, Tobin MD, Silverman EK, Boezen HM, Cho MH, COPDGene Investigators, ECLIPSE Investigators, LifeLines Investigators, SPIROMICS Research Group, International COPD Genetics Network Investigators, UK BiLEVE Investigators, International COPD Genetics Consortium. Genetic loci associated with chronic obstructive pulmonary disease overlap with loci for lung function and pulmonary fibrosis. Nat Genet 2017; 49:426-432. [PMID: 28166215 PMCID: PMC5381275 DOI: 10.1038/ng.3752] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/23/2016] [Indexed: 12/15/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of mortality worldwide. We performed a genetic association study in 15,256 cases and 47,936 controls, with replication of select top results (P < 5 × 10-6) in 9,498 cases and 9,748 controls. In the combined meta-analysis, we identified 22 loci associated at genome-wide significance, including 13 new associations with COPD. Nine of these 13 loci have been associated with lung function in general population samples, while 4 (EEFSEC, DSP, MTCL1, and SFTPD) are new. We noted two loci shared with pulmonary fibrosis (FAM13A and DSP) but that had opposite risk alleles for COPD. None of our loci overlapped with genome-wide associations for asthma, although one locus has been implicated in joint susceptibility to asthma and obesity. We also identified genetic correlation between COPD and asthma. Our findings highlight new loci associated with COPD, demonstrate the importance of specific loci associated with lung function to COPD, and identify potential regions of genetic overlap between COPD and other respiratory diseases.
Collapse
Affiliation(s)
- Brian D. Hobbs
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and
Women’s Hospital, Boston, MA, USA
| | - Kim de Jong
- University of Groningen, University Medical Center Groningen,
Department of Epidemiology, Groningen, the Netherlands
- University of Groningen, University Medical Center Groningen,
Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the
Netherlands
| | - Maxime Lamontagne
- Institut universitaire de cardiologie et de pneumologie de
Québec, Québec, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de
Québec, Québec, Canada
- Department of Molecular Medicine, Laval University, Québec,
Canada
| | - Nick Shrine
- Genetic Epidemiology Group, Department of Health Sciences,
University of Leicester, Leicester, UK
| | - María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences,
University of Leicester, Leicester, UK
| | - Louise V. Wain
- Genetic Epidemiology Group, Department of Health Sciences,
University of Leicester, Leicester, UK
| | - Ian P. Hall
- Division of Respiratory Medicine, Queen’s Medical Centre,
University of Nottingham, Nottingham, UK
| | - Victoria E. Jackson
- Genetic Epidemiology Group, Department of Health Sciences,
University of Leicester, Leicester, UK
| | - Annah B. Wyss
- Epidemiology Branch, National Institute of Environmental Health
Sciences, National Institutes of Health, Department of Health and Human Services,
Research Triangle Park, NC, USA
| | - Stephanie J. London
- Epidemiology Branch, National Institute of Environmental Health
Sciences, National Institutes of Health, Department of Health and Human Services,
Research Triangle Park, NC, USA
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel
Hill, NC, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel
Hill, NC, USA
| | - David P. Strachan
- Population Health Research Institute, St. George’s,
University of London, London, UK
| | - Terri H. Beaty
- Johns Hopkins University Bloomberg School of Public Health,
Baltimore, MD, USA
| | - John E. Hokanson
- Department of Epidemiology, University of Colorado Anschutz Medical
Campus, Aurora, CO, USA
| | - James D. Crapo
- Department of Medicine, Division of Pulmonary and Critical Care
Medicine, National Jewish Health, Denver, CO, USA
| | - Peter J. Castaldi
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
- Division of General Internal Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
| | - Robert P. Chase
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, University of Washington,
Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA,
USA
- Department of Biostatistics, University of Washington, Seattle, WA,
USA
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit, University of Washington,
Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA,
USA
- Group Health Research Institute, Group Health Cooperative, Seattle,
WA, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington,
Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA,
USA
- Department of Epidemiology, University of Washington, Seattle, WA,
USA
- Group Health Research Institute, Group Health Cooperative, Seattle,
WA, USA
- Department of Health Services, University of Washington, Seattle,
WA, USA
| | - Sina A. Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine
Sleep Center, Department of Medicine, University of Washington, Seattle, WA,
USA
| | - Pieter Zanen
- Department of Pulmonology, University Medical Center Utrecht,
University of Utrecht, Utrecht, the Netherlands
| | - Jan W. Lammers
- Department of Pulmonology, University Medical Center Utrecht,
University of Utrecht, Utrecht, the Netherlands
| | - Matthijs Oudkerk
- University of Groningen, University Medical Center Groningen,
Center for Medical Imaging, the Netherlands
| | - H. J. Groen
- University of Groningen, University Medical Center Groningen,
Department of Pulmonology, Groningen, the Netherlands
| | | | | | - Stephen I. Rennard
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of
Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Clinical Discovery Unit, AstraZeneca, Cambridge, UK
| | - Jørgen Vestbo
- School of Biological Sciences, University of Manchester,
Manchester, UK
| | - Wim Timens
- Department of Pathology and Medical Biology, University of
Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen,
the Netherlands
| | - Peter D. Paré
- University of British Columbia Center for Heart Lung Innovation and
Institute for Heart and Lung Health, St Paul’s Hospital, Vancouver, British
Columbia, Canada
| | | | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public
Health, Boston, MA, USA
- The National Heart, Lung, and Blood Institute’s Framingham
Heart Study, Framingham, MA, USA
| | - George T. O’Connor
- The National Heart, Lung, and Blood Institute’s Framingham
Heart Study, Framingham, MA, USA
- Pulmonary Center, Department of Medicine, Boston University School
of Medicine, Boston, MA, USA
| | - Jemma B. Wilk
- The National Heart, Lung, and Blood Institute’s Framingham
Heart Study, Framingham, MA, USA
| | - Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center,
School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Mi Kyeong Lee
- Department of Internal Medicine and Environmental Health Center,
School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, and Clinical
Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center,
University of Ulsan College of Medicine, Seoul, South Korea
| | - Judith M. Vonk
- University of Groningen, University Medical Center Groningen,
Department of Epidemiology, Groningen, the Netherlands
- University of Groningen, University Medical Center Groningen,
Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the
Netherlands
| | - Harry J. de Koning
- Department of Public Health, Erasmus Medical Center Rotterdam,
Rotterdam, the Netherlands
| | - Shuguang Leng
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | | | | | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia,
Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia,
Charlottesville, VA, USA
| | - Xin-Qun Wang
- Department of Public Health Sciences, University of Virginia,
Charlottesville, VA, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia,
Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia,
Charlottesville, VA, USA
| | - R Graham Barr
- Department of Medicine, College of Physicians and Surgeons and
Department of Epidemiology, Mailman School of Public Health, Columbia University,
New York, NY, USA
| | - David Sparrow
- VA Boston Healthcare System and Department of Medicine, Boston
University School of Medicine, Boston, MA, USA
| | - Augusto A. Litonjua
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and
Women’s Hospital, Boston, MA, USA
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen,
Norway
| | - Amund Gulsvik
- Department of Clinical Science, University of Bergen, Bergen,
Norway
| | - Lies Lahousse
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the
Netherlands
- Department of Respiratory Medicine, Ghent University Hospital,
Ghent, Belgium
| | - Guy G. Brusselle
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the
Netherlands
- Department of Respiratory Medicine, Ghent University Hospital,
Ghent, Belgium
- Department of Respiratory Medicine, Erasmus Medical Center,
Rotterdam, the Netherlands
| | - Bruno H. Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the
Netherlands
- Netherlands Health Care Inspectorate, The Hague, the
Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam,
the Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Leiden, the Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the
Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam,
the Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Leiden, the Netherlands
| | - Elizabeth J. Ampleford
- Center for Genomics and Personalized Medicine Research, Wake Forest
University School of Medicine, Winston Salem, NC, USA
| | - Eugene R. Bleecker
- Center for Genomics and Personalized Medicine Research, Wake Forest
University School of Medicine, Winston Salem, NC, USA
| | - Prescott G. Woodruff
- Cardiovascular Research Institute and the Department of Medicine,
Division of Pulmonary, Critical Care, Sleep, and Allergy, University of California
at San Francisco, San Francisco, CA, USA
| | - Deborah A. Meyers
- Center for Genomics and Personalized Medicine Research, Wake Forest
University School of Medicine, Winston Salem, NC, USA
| | - Dandi Qiao
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
| | | | - Jae-Joon Yim
- Division of Pulmonary and Critical Care Medicine, Department of
Internal Medicine, Seoul National University College of Medicine, Seoul, South
Korea
| | - Deog Kyeom Kim
- Seoul National University College of Medicine, SMG-SNU Boramae
Medical Center, Seoul, South Korea
| | - Iwona Hawrylkiewicz
- 2nd Department of Respiratory Medicine, Institute of Tuberculosis
and Lung Diseases, Warsaw, Poland
| | - Pawel Sliwinski
- 2nd Department of Respiratory Medicine, Institute of Tuberculosis
and Lung Diseases, Warsaw, Poland
| | - Megan Hardin
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and
Women’s Hospital, Boston, MA, USA
- Clinical Discovery Unit, AstraZeneca, Cambridge, UK
| | - Tasha E. Fingerlin
- Center for Genes, Environment and Health, National Jewish Health,
Denver, CO, USA
- Department of Biostatistics and Informatics, University of Colorado
Denver, Aurora, CO, USA
| | - David A. Schwartz
- Center for Genes, Environment and Health, National Jewish Health,
Denver, CO, USA
- Department of Medicine, School of Medicine, University of Colorado
Denver, Aurora, CO, USA
- Department of Immunology, School of Medicine, University of
Colorado Denver, Aurora, CO, USA
| | - Dirkje S. Postma
- University of Groningen, University Medical Center Groningen,
Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the
Netherlands
- University of Groningen, University Medical Center Groningen,
Department of Pulmonology, Groningen, the Netherlands
| | | | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Health Sciences,
University of Leicester, Leicester, UK
- National Institute for Health Research (NIHR) Leicester Respiratory
Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and
Women’s Hospital, Boston, MA, USA
| | - H. Marike Boezen
- University of Groningen, University Medical Center Groningen,
Department of Epidemiology, Groningen, the Netherlands
- University of Groningen, University Medical Center Groningen,
Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the
Netherlands
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s
Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and
Women’s Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
966
|
Pacifico R, Davis RL. Transcriptome sequencing implicates dorsal striatum-specific gene network, immune response and energy metabolism pathways in bipolar disorder. Mol Psychiatry 2017; 22:441-449. [PMID: 27350034 DOI: 10.1038/mp.2016.94] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 04/08/2016] [Accepted: 05/09/2016] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a highly heritable and heterogeneous mental illness whose manifestations often include impulsive and risk-taking behavior. This particular phenotype suggests that abnormal striatal function could be involved in BD etiology, yet most transcriptomic studies of this disorder have concentrated on cortical brain regions. We believe we report the first transcriptome sequencing of the postmortem human dorsal striatum comparing bipolar (18) and control (17) subjects. Fourteen genes were detected as differentially expressed at a 5% false discovery rate, including a few immune response genes such as NLRC5, S100A12, LILRA4 and FCGBP, as well as an assortment of non-protein coding genes. Functional pathway analysis found an enrichment of upregulated genes across many immune/inflammation pathways and an enrichment of downregulated genes among oxidative phosphorylation pathways. Co-expression network analysis revealed 20 modules of highly interconnected genes; two of the modules were significantly enriched for BD susceptibility single-nucleotide polymorphisms deriving from a large genome-wide association study data set. Remarkably, the module with the highest genetic association signal for BD, which contained many genes from signaling pathways, was also enriched in markers characteristic of gene expression in dorsal striatum medium spiny neurons-unlike most other modules, which showed no such regional and neuronal specificity. These findings draw a link between BD etiology at the gene level and a specific brain region, and highlight striatal signaling pathways as potential targets for the development of novel treatments to manage BD.
Collapse
Affiliation(s)
- R Pacifico
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, USA
| | - R L Davis
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, USA
| |
Collapse
|
967
|
Trampush JW, Yang MLZ, Yu J, Knowles E, Davies G, Liewald DC, Starr JM, Djurovic S, Melle I, Sundet K, Christoforou A, Reinvang I, DeRosse P, Lundervold AJ, Steen VM, Espeseth T, Räikkönen K, Widen E, Palotie A, Eriksson JG, Giegling I, Konte B, Roussos P, Giakoumaki S, Burdick KE, Payton A, Ollier W, Horan M, Chiba-Falek O, Attix DK, Need AC, Cirulli ET, Voineskos AN, Stefanis NC, Avramopoulos D, Hatzimanolis A, Arking DE, Smyrnis N, Bilder RM, Freimer NA, Cannon TD, London E, Poldrack RA, Sabb FW, Congdon E, Conley ED, Scult MA, Dickinson D, Straub RE, Donohoe G, Morris D, Corvin A, Gill M, Hariri AR, Weinberger DR, Pendleton N, Bitsios P, Rujescu D, Lahti J, Le Hellard S, Keller MC, Andreassen OA, Deary IJ, Glahn DC, Malhotra AK, Lencz T. GWAS meta-analysis reveals novel loci and genetic correlates for general cognitive function: a report from the COGENT consortium. Mol Psychiatry 2017; 22:336-345. [PMID: 28093568 PMCID: PMC5322272 DOI: 10.1038/mp.2016.244] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/30/2016] [Accepted: 11/03/2016] [Indexed: 01/12/2023]
Abstract
The complex nature of human cognition has resulted in cognitive genomics lagging behind many other fields in terms of gene discovery using genome-wide association study (GWAS) methods. In an attempt to overcome these barriers, the current study utilized GWAS meta-analysis to examine the association of common genetic variation (~8M single-nucleotide polymorphisms (SNP) with minor allele frequency ⩾1%) to general cognitive function in a sample of 35 298 healthy individuals of European ancestry across 24 cohorts in the Cognitive Genomics Consortium (COGENT). In addition, we utilized individual SNP lookups and polygenic score analyses to identify genetic overlap with other relevant neurobehavioral phenotypes. Our primary GWAS meta-analysis identified two novel SNP loci (top SNPs: rs76114856 in the CENPO gene on chromosome 2 and rs6669072 near LOC105378853 on chromosome 1) associated with cognitive performance at the genome-wide significance level (P<5 × 10-8). Gene-based analysis identified an additional three Bonferroni-corrected significant loci at chromosomes 17q21.31, 17p13.1 and 1p13.3. Altogether, common variation across the genome resulted in a conservatively estimated SNP heritability of 21.5% (s.e.=0.01%) for general cognitive function. Integration with prior GWAS of cognitive performance and educational attainment yielded several additional significant loci. Finally, we found robust polygenic correlations between cognitive performance and educational attainment, several psychiatric disorders, birth length/weight and smoking behavior, as well as a novel genetic association to the personality trait of openness. These data provide new insight into the genetics of neurocognitive function with relevance to understanding the pathophysiology of neuropsychiatric illness.
Collapse
Affiliation(s)
- J W Trampush
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA
| | - M L Z Yang
- Institute of Mental Health, Singapore, Singapore
| | - J Yu
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA,Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - E Knowles
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - G Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - D C Liewald
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - J M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - S Djurovic
- Department of Medical Genetics, Oslo University Hospital, University of Bergen, Oslo, Norway,NORMENT, K.G. Jebsen Centre for Psychosis Research, University of Bergen, Bergen, Norway
| | - I Melle
- NORMENT, K.G. Jebsen Centre for Psychosis Research, University of Bergen, Bergen, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - K Sundet
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Psychology, University of Oslo, Oslo, Norway
| | - A Christoforou
- NORMENT, K.G. Jebsen Centre for Psychosis Research, University of Bergen, Bergen, Norway,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - I Reinvang
- Department of Psychology, University of Oslo, Oslo, Norway
| | - P DeRosse
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA,Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - A J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - V M Steen
- NORMENT, K.G. Jebsen Centre for Psychosis Research, University of Bergen, Bergen, Norway,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - T Espeseth
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Department of Psychology, University of Oslo, Oslo, Norway
| | - K Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - E Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - A Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK,Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - J G Eriksson
- National Institute for Health and Welfare, Helsinki, Finland,Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland,Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland,Folkhälsan Research Centre, Helsinki, Finland
| | - I Giegling
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - B Konte
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - P Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Mental Illness Research, Education, and Clinical Center (VISN 3), James J. Peters VA Medical Center, Bronx, NY, USA
| | - S Giakoumaki
- Department of Psychology, University of Crete, Rethymno, Greece
| | - K E Burdick
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Mental Illness Research, Education, and Clinical Center (VISN 3), James J. Peters VA Medical Center, Bronx, NY, USA
| | - A Payton
- Manchester Centre for Audiology and Deafness, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK,Division of Evolution and Genomic Sciences, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - W Ollier
- Centre for Integrated Genomic Medical Research, Institute of Population Health, University of Manchester, Manchester, UK
| | - M Horan
- Manchester Medical School, Institute of Brain, Behaviour, and Mental Health, University of Manchester, Manchester, UK
| | - O Chiba-Falek
- Department of Neurology, Bryan Alzheimer's Disease Research Center, and Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - D K Attix
- Department of Neurology, Bryan Alzheimer's Disease Research Center, and Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA,Division of Medical Psychology, Department of Neurology, Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - A C Need
- Division of Brain Sciences, Department of Medicine, Imperial College, London, UK
| | - E T Cirulli
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA
| | - A N Voineskos
- Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - N C Stefanis
- Department of Psychiatry, University of Athens School of Medicine, Eginition Hospital, Athens, Greece,University Mental Health Research Institute, Athens, Greece,Neurobiology Research Institute, Theodor Theohari Cozzika Foundation, Athens, Greece
| | - D Avramopoulos
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Psychiatry and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - A Hatzimanolis
- Department of Psychiatry, University of Athens School of Medicine, Eginition Hospital, Athens, Greece,University Mental Health Research Institute, Athens, Greece,Neurobiology Research Institute, Theodor Theohari Cozzika Foundation, Athens, Greece
| | - D E Arking
- Department of Psychiatry and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - N Smyrnis
- Department of Psychiatry, University of Athens School of Medicine, Eginition Hospital, Athens, Greece,University Mental Health Research Institute, Athens, Greece
| | - R M Bilder
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - N A Freimer
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - T D Cannon
- Department of Psychology, Yale University, New Haven, CT, USA
| | - E London
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - R A Poldrack
- Department of Psychology, Stanford University, Palo Alto, CA, USA
| | - F W Sabb
- Robert and Beverly Lewis Center for Neuroimaging, University of Oregon, Eugene, OR, USA
| | - E Congdon
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | | | - M A Scult
- Department of Psychology & Neuroscience, Laboratory of NeuroGenetics, Duke University, Durham, NC, USA
| | - D Dickinson
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institute of Health, Bethesda, MD, USA
| | - R E Straub
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - G Donohoe
- Department of Psychology, National University of Ireland, Galway, Ireland
| | - D Morris
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - A Corvin
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - M Gill
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - A R Hariri
- Department of Psychology & Neuroscience, Laboratory of NeuroGenetics, Duke University, Durham, NC, USA
| | - D R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - N Pendleton
- Centre for Integrated Genomic Medical Research, Institute of Population Health, University of Manchester, Manchester, UK,Manchester Medical School, Institute of Brain, Behaviour, and Mental Health, University of Manchester, Manchester, UK
| | - P Bitsios
- Department of Psychiatry and Behavioral Sciences, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - D Rujescu
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - J Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland,Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
| | - S Le Hellard
- NORMENT, K.G. Jebsen Centre for Psychosis Research, University of Bergen, Bergen, Norway,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - M C Keller
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - O A Andreassen
- NORMENT, K.G. Jebsen Centre for Psychosis Research, University of Bergen, Bergen, Norway,Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - I J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - D C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - A K Malhotra
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA,Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA,Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, NY, USA
| | - T Lencz
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA,Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA,Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, NY, USA,Division of Psychiatry Research, Zucker Hillside Hospital, 75-59 263rd Street, Glen Oaks, NY 11004, USA. E-mail:
| |
Collapse
|
968
|
Pulit SL, Karaderi T, Lindgren CM. Sexual dimorphisms in genetic loci linked to body fat distribution. Biosci Rep 2017; 37:BSR20160184. [PMID: 28073971 PMCID: PMC5291139 DOI: 10.1042/bsr20160184] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/07/2017] [Accepted: 01/10/2017] [Indexed: 01/02/2023] Open
Abstract
Obesity is a chronic condition associated with increased morbidity and mortality and is a risk factor for a number of other diseases including type 2 diabetes and cardiovascular disease. Obesity confers an enormous, costly burden on both individuals and public health more broadly. Body fat distribution is a heritable trait and a well-established predictor of adverse metabolic outcomes. Body fat distribution is distinct from overall obesity in measurement, but studies of body fat distribution can yield insights into the risk factors for and causes of overall obesity. Sexual dimorphism in body fat distribution is present throughout life. Though sexual dimorphism is subtle in early stages of life, it is attenuated in puberty and during menopause. This phenomenon could be, at least in part, due to the influence of sex hormones on the trait. Findings from recent large genome-wide association studies (GWAS) for various measures of body fat distribution (including waist-to-hip ratio, hip or waist circumference, trunk fat percentage and the ratio of android and gynoid fat percentage) emphasize the strong sexual dimorphism in the genetic regulation of fat distribution traits. Importantly, sexual dimorphism is not observed for overall obesity (as assessed by body mass index or total fat percentage). Notably, the genetic loci associated with body fat distribution, which show sexual dimorphism, are located near genes that are expressed in adipose tissues and/or adipose cells. Considering the epidemiological and genetic evidence, sexual dimorphism is a prominent feature of body fat distribution. Research that specifically focuses on sexual dimorphism in fat distribution can provide novel insights into human physiology and into the development of obesity and its comorbidities, as well as yield biological clues that will aid in the improvement of disease prevention and treatment.
Collapse
Affiliation(s)
- Sara L Pulit
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tugce Karaderi
- Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, Famagusta, Cyprus
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, U.K.
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| |
Collapse
|
969
|
Diversity in non-repetitive human sequences not found in the reference genome. Nat Genet 2017; 49:588-593. [PMID: 28250455 DOI: 10.1038/ng.3801] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/03/2017] [Indexed: 12/15/2022]
Abstract
Genomes usually contain some non-repetitive sequences that are missing from the reference genome and occur only in a population subset. Such non-repetitive, non-reference (NRNR) sequences have remained largely unexplored in terms of their characterization and downstream analyses. Here we describe 3,791 breakpoint-resolved NRNR sequence variants called using PopIns from whole-genome sequence data of 15,219 Icelanders. We found that over 95% of the 244 NRNR sequences that are 200 bp or longer are present in chimpanzees, indicating that they are ancestral. Furthermore, 149 variant loci are in linkage disequilibrium (r2 > 0.8) with a genome-wide association study (GWAS) catalog marker, suggesting disease relevance. Additionally, we report an association (P = 3.8 × 10-8, odds ratio (OR) = 0.92) with myocardial infarction (23,360 cases, 300,771 controls) for a 766-bp NRNR sequence variant. Our results underline the importance of including variation of all complexity levels when searching for variants that associate with disease.
Collapse
|
970
|
McCoy RC, Wakefield J, Akey JM. Impacts of Neanderthal-Introgressed Sequences on the Landscape of Human Gene Expression. Cell 2017; 168:916-927.e12. [PMID: 28235201 PMCID: PMC6219754 DOI: 10.1016/j.cell.2017.01.038] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/09/2017] [Accepted: 01/27/2017] [Indexed: 11/20/2022]
Abstract
Regulatory variation influencing gene expression is a key contributor to phenotypic diversity, both within and between species. Unfortunately, RNA degrades too rapidly to be recovered from fossil remains, limiting functional genomic insights about our extinct hominin relatives. Many Neanderthal sequences survive in modern humans due to ancient hybridization, providing an opportunity to assess their contributions to transcriptional variation and to test hypotheses about regulatory evolution. We developed a flexible Bayesian statistical approach to quantify allele-specific expression (ASE) in complex RNA-seq datasets. We identified widespread expression differences between Neanderthal and modern human alleles, indicating pervasive cis-regulatory impacts of introgression. Brain regions and testes exhibited significant downregulation of Neanderthal alleles relative to other tissues, consistent with natural selection influencing the tissue-specific regulatory landscape. Our study demonstrates that Neanderthal-inherited sequences are not silent remnants of ancient interbreeding but have measurable impacts on gene expression that contribute to variation in modern human phenotypes.
Collapse
Affiliation(s)
- Rajiv C McCoy
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jon Wakefield
- Department of Statistics, University of Washington, Seattle, WA 98195, USA; Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Joshua M Akey
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
971
|
Zemel BS. Influence of complex childhood diseases on variation in growth and skeletal development. Am J Hum Biol 2017; 29. [DOI: 10.1002/ajhb.22985] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 01/22/2017] [Accepted: 02/06/2017] [Indexed: 01/10/2023] Open
Affiliation(s)
- Babette S. Zemel
- Division of Gastroenterology, Hepatology and Nutrition; The Children's Hospital of Philadelphia, Philadelphia; Pennsylvania
- Department of Pediatrics; The University of Pennsylvania Perelman School of Medicine, Philadelphia; Pennsylvania
| |
Collapse
|
972
|
Sequence variant at 8q24.21 associates with sciatica caused by lumbar disc herniation. Nat Commun 2017; 8:14265. [PMID: 28223688 PMCID: PMC5322534 DOI: 10.1038/ncomms14265] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 12/14/2016] [Indexed: 12/19/2022] Open
Abstract
Lumbar disc herniation (LDH) is common and often debilitating. Microdiscectomy of herniated lumbar discs (LDHsurg) is performed on the most severe cases to resolve the resulting sciatica. Here we perform a genome-wide association study on 4,748 LDHsurg cases and 282,590 population controls and discover 37 highly correlated markers associating with LDHsurg at 8q24.21 (between CCDC26 and GSDMC), represented by rs6651255[C] (OR=0.81; P=5.6 × 10−12) with a stronger effect among younger patients than older. As rs6651255[C] also associates with height, we performed a Mendelian randomization analysis using height polygenic risk scores as instruments to estimate the effect of height on LDHsurg risk, and found that the marker's association with LDHsurg is much greater than predicted by its effect on height. In light of presented findings, we speculate that the effect of rs6651255 on LDHsurg is driven by susceptibility to developing severe and persistent sciatica upon LDH. Lumbar disc herniation (LDH) can cause persistent sciatica, and in some cases surgery is required to relieve symptoms. Here, the authors carry out a genome-wide association study using microdiscectomy as an indicator of severe LDH, and find a locus on chromosome 8 associated with this condition.
Collapse
|
973
|
Fava VM, Manry J, Cobat A, Orlova M, Van Thuc N, Moraes MO, Sales-Marques C, Stefani MMA, Latini ACP, Belone AF, Thai VH, Abel L, Alcaïs A, Schurr E. A genome wide association study identifies a lncRna as risk factor for pathological inflammatory responses in leprosy. PLoS Genet 2017; 13:e1006637. [PMID: 28222097 PMCID: PMC5340414 DOI: 10.1371/journal.pgen.1006637] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 03/07/2017] [Accepted: 02/14/2017] [Indexed: 01/23/2023] Open
Abstract
Leprosy Type-1 Reactions (T1Rs) are pathological inflammatory responses that afflict a sub-group of leprosy patients and result in peripheral nerve damage. Here, we employed a family-based GWAS in 221 families with 229 T1R-affect offspring with stepwise replication to identify risk factors for T1R. We discovered, replicated and validated T1R-specific associations with SNPs located in chromosome region 10p21.2. Combined analysis across the three independent samples resulted in strong evidence of association of rs1875147 with T1R (p = 4.5x10-8; OR = 1.54, 95% CI = 1.32–1.80). The T1R-risk locus was restricted to a lncRNA-encoding genomic interval with rs1875147 being an eQTL for the lncRNA. Since a genetic overlap between leprosy and inflammatory bowel disease (IBD) has been detected, we evaluated if the shared genetic control could be traced to the T1R endophenotype. Employing the results of a recent IBD GWAS meta-analysis we found that 10.6% of IBD SNPs available in our dataset shared a common risk-allele with T1R (p = 2.4x10-4). This finding points to a substantial overlap in the genetic control of clinically diverse inflammatory disorders. Leprosy still affects approximately 200,000 new victims each year. A major challenge of leprosy control is the prevention of permanent disability due to nerve damage. Nerve damage occurs if leprosy remains undiagnosed for extended periods or when patients undergo pathological inflammatory responses termed Type-1 Reactions (T1R). T1R is a rare example where beneficial inflammatory responses are temporal separated from host pathological responses. There is strong experimental evidence that supports a role of host genetic factors in T1R susceptibility. Here, we employed a genome-wide association study (GWAS) to investigate susceptibility factors for T1R in Vietnamese families. We followed up the initial GWAS findings in independent population samples from Vietnam and Brazil and identified a set of cis-eQTL genetic variants for the ENSG00000235140 lncRNA as global risk factors for T1R. To test our proposal that T1R is a strong model for pathological inflammatory responses we evaluated if inflammatory bowel disease (IBD) genetic risk-factors were enriched among T1R risk factors. We observed that more than 10% of IBD-risk loci were nominally associated with risk for T1R suggesting a shared mechanism of excessive inflammatory response in the both disease etiologies.
Collapse
Affiliation(s)
- Vinicius M. Fava
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada
- * E-mail: (ES); (VMF)
| | - Jeremy Manry
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- University Paris Descartes, Imagine Institute, Paris, France
- Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, United States of America
| | - Marianna Orlova
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada
| | | | - Milton O. Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Mariane M. A. Stefani
- Tropical Pathology and Public Health Institute, Federal University of Goiás, Goiânia, Brazil
| | | | | | - Vu Hong Thai
- Hospital for Dermato-Venerology, Ho Chi Minh City, Vietnam
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- University Paris Descartes, Imagine Institute, Paris, France
- Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, United States of America
| | - Alexandre Alcaïs
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- University Paris Descartes, Imagine Institute, Paris, France
- Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, United States of America
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The McGill International TB Centre, Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada
- * E-mail: (ES); (VMF)
| |
Collapse
|
974
|
Lampl M, Schoen M. How long bones grow children: Mechanistic paths to variation in human height growth. Am J Hum Biol 2017; 29. [DOI: 10.1002/ajhb.22983] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/01/2017] [Accepted: 02/05/2017] [Indexed: 12/25/2022] Open
Affiliation(s)
- Michelle Lampl
- Center for the Study of Human Health; Emory University; Atlanta Georgia 30324
- Department of Anthropology; Emory University; Atlanta Georgia 30324
| | - Meriah Schoen
- Center for the Study of Human Health; Emory University; Atlanta Georgia 30324
- Department of Nutrition; Georgia State University; Atlanta Georgia 30302
| |
Collapse
|
975
|
Chung D, Kim HJ, Zhao H. graph-GPA: A graphical model for prioritizing GWAS results and investigating pleiotropic architecture. PLoS Comput Biol 2017; 13:e1005388. [PMID: 28212402 PMCID: PMC5347371 DOI: 10.1371/journal.pcbi.1005388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 03/06/2017] [Accepted: 01/28/2017] [Indexed: 02/06/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified tens of thousands of genetic variants associated with hundreds of phenotypes and diseases, which have provided clinical and medical benefits to patients with novel biomarkers and therapeutic targets. However, identification of risk variants associated with complex diseases remains challenging as they are often affected by many genetic variants with small or moderate effects. There has been accumulating evidence suggesting that different complex traits share common risk basis, namely pleiotropy. Recently, several statistical methods have been developed to improve statistical power to identify risk variants for complex traits through a joint analysis of multiple GWAS datasets by leveraging pleiotropy. While these methods were shown to improve statistical power for association mapping compared to separate analyses, they are still limited in the number of phenotypes that can be integrated. In order to address this challenge, in this paper, we propose a novel statistical framework, graph-GPA, to integrate a large number of GWAS datasets for multiple phenotypes using a hidden Markov random field approach. Application of graph-GPA to a joint analysis of GWAS datasets for 12 phenotypes shows that graph-GPA improves statistical power to identify risk variants compared to statistical methods based on smaller number of GWAS datasets. In addition, graph-GPA also promotes better understanding of genetic mechanisms shared among phenotypes, which can potentially be useful for the development of improved diagnosis and therapeutics. The R implementation of graph-GPA is currently available at https://dongjunchung.github.io/GGPA/. Recently, there has been accumulating evidence suggesting pleiotropy, i.e., genetic components shared across multiple phenotypes. Incorporation of pleiotropy in genetic analysis might improve statistical power to identify risk associated genetic variants. Several statistical approaches have been proposed to utilize pleiotropy for association mapping but they are currently still limited to a relatively small number of phenotypes, e.g., a pair of phenotypes. This restricts potential gain in statistical power in association mapping and investigation of pleiotropic structure among a large number of phenotypes. In order to address this challenge, in this paper, we propose graph-GPA, a novel statistical framework to integrate a large number of phenotypes using a hidden Markov random field architecture. Application of the proposed statistical method to GWAS datasets for 12 phenotypes showed that graph-GPA does not only provide a parsimonious representation of genetic relationship among these phenotypes, but also identify significantly larger number of novel genetic variants that are potentially functional. We believe that this novel approach might help investigation of common etiology and improvement of diagnosis and therapeutics.
Collapse
Affiliation(s)
- Dongjun Chung
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| | - Hang J. Kim
- Department of Mathematical Sciences, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, United States of America
- VA Cooperative Studies Program Coordinating Center, West Haven, Connecticut, United States of America
| |
Collapse
|
976
|
Marouli E, Graff M, Medina-Gomez C, Lo KS, Wood AR, Kjaer TR, Fine RS, Lu Y, Schurmann C, Highland HM, Rüeger S, Thorleifsson G, Justice AE, Lamparter D, Stirrups KE, Turcot V, Young KL, Winkler TW, Esko T, Karaderi T, Locke AE, Masca NGD, Ng MCY, Mudgal P, Rivas MA, Vedantam S, Mahajan A, Guo X, Abecasis G, Aben KK, Adair LS, Alam DS, Albrecht E, Allin KH, Allison M, Amouyel P, Appel EV, Arveiler D, Asselbergs FW, Auer PL, Balkau B, Banas B, Bang LE, Benn M, Bergmann S, Bielak LF, Blüher M, Boeing H, Boerwinkle E, Böger CA, Bonnycastle LL, Bork-Jensen J, Bots ML, Bottinger EP, Bowden DW, Brandslund I, Breen G, Brilliant MH, Broer L, Burt AA, Butterworth AS, Carey DJ, Caulfield MJ, Chambers JC, Chasman DI, Chen YDI, Chowdhury R, Christensen C, Chu AY, Cocca M, Collins FS, Cook JP, Corley J, Galbany JC, Cox AJ, Cuellar-Partida G, Danesh J, Davies G, de Bakker PIW, de Borst GJ, de Denus S, de Groot MCH, de Mutsert R, Deary IJ, Dedoussis G, Demerath EW, den Hollander AI, Dennis JG, Di Angelantonio E, Drenos F, Du M, Dunning AM, Easton DF, Ebeling T, Edwards TL, Ellinor PT, Elliott P, Evangelou E, Farmaki AE, Faul JD, et alMarouli E, Graff M, Medina-Gomez C, Lo KS, Wood AR, Kjaer TR, Fine RS, Lu Y, Schurmann C, Highland HM, Rüeger S, Thorleifsson G, Justice AE, Lamparter D, Stirrups KE, Turcot V, Young KL, Winkler TW, Esko T, Karaderi T, Locke AE, Masca NGD, Ng MCY, Mudgal P, Rivas MA, Vedantam S, Mahajan A, Guo X, Abecasis G, Aben KK, Adair LS, Alam DS, Albrecht E, Allin KH, Allison M, Amouyel P, Appel EV, Arveiler D, Asselbergs FW, Auer PL, Balkau B, Banas B, Bang LE, Benn M, Bergmann S, Bielak LF, Blüher M, Boeing H, Boerwinkle E, Böger CA, Bonnycastle LL, Bork-Jensen J, Bots ML, Bottinger EP, Bowden DW, Brandslund I, Breen G, Brilliant MH, Broer L, Burt AA, Butterworth AS, Carey DJ, Caulfield MJ, Chambers JC, Chasman DI, Chen YDI, Chowdhury R, Christensen C, Chu AY, Cocca M, Collins FS, Cook JP, Corley J, Galbany JC, Cox AJ, Cuellar-Partida G, Danesh J, Davies G, de Bakker PIW, de Borst GJ, de Denus S, de Groot MCH, de Mutsert R, Deary IJ, Dedoussis G, Demerath EW, den Hollander AI, Dennis JG, Di Angelantonio E, Drenos F, Du M, Dunning AM, Easton DF, Ebeling T, Edwards TL, Ellinor PT, Elliott P, Evangelou E, Farmaki AE, Faul JD, Feitosa MF, Feng S, Ferrannini E, Ferrario MM, Ferrieres J, Florez JC, Ford I, Fornage M, Franks PW, Frikke-Schmidt R, Galesloot TE, Gan W, Gandin I, Gasparini P, Giedraitis V, Giri A, Girotto G, Gordon SD, Gordon-Larsen P, Gorski M, Grarup N, Grove ML, Gudnason V, Gustafsson S, Hansen T, Harris KM, Harris TB, Hattersley AT, Hayward C, He L, Heid IM, Heikkilä K, Helgeland Ø, Hernesniemi J, Hewitt AW, Hocking LJ, Hollensted M, Holmen OL, Hovingh GK, Howson JMM, Hoyng CB, Huang PL, Hveem K, Ikram MA, Ingelsson E, Jackson AU, Jansson JH, Jarvik GP, Jensen GB, Jhun MA, Jia Y, Jiang X, Johansson S, Jørgensen ME, Jørgensen T, Jousilahti P, Jukema JW, Kahali B, Kahn RS, Kähönen M, Kamstrup PR, Kanoni S, Kaprio J, Karaleftheri M, Kardia SLR, Karpe F, Kee F, Keeman R, Kiemeney LA, Kitajima H, Kluivers KB, Kocher T, Komulainen P, Kontto J, Kooner JS, Kooperberg C, Kovacs P, Kriebel J, Kuivaniemi H, Küry S, Kuusisto J, La Bianca M, Laakso M, Lakka TA, Lange EM, Lange LA, Langefeld CD, Langenberg C, Larson EB, Lee IT, Lehtimäki T, Lewis CE, Li H, Li J, Li-Gao R, Lin H, Lin LA, Lin X, Lind L, Lindström J, Linneberg A, Liu Y, Liu Y, Lophatananon A, Luan J, Lubitz SA, Lyytikäinen LP, Mackey DA, Madden PAF, Manning AK, Männistö S, Marenne G, Marten J, Martin NG, Mazul AL, Meidtner K, Metspalu A, Mitchell P, Mohlke KL, Mook-Kanamori DO, Morgan A, Morris AD, Morris AP, Müller-Nurasyid M, Munroe PB, Nalls MA, Nauck M, Nelson CP, Neville M, Nielsen SF, Nikus K, Njølstad PR, Nordestgaard BG, Ntalla I, O'Connel JR, Oksa H, Loohuis LMO, Ophoff RA, Owen KR, Packard CJ, Padmanabhan S, Palmer CNA, Pasterkamp G, Patel AP, Pattie A, Pedersen O, Peissig PL, Peloso GM, Pennell CE, Perola M, Perry JA, Perry JRB, Person TN, Pirie A, Polasek O, Posthuma D, Raitakari OT, Rasheed A, Rauramaa R, Reilly DF, Reiner AP, Renström F, Ridker PM, Rioux JD, Robertson N, Robino A, Rolandsson O, Rudan I, Ruth KS, Saleheen D, Salomaa V, Samani NJ, Sandow K, Sapkota Y, Sattar N, Schmidt MK, Schreiner PJ, Schulze MB, Scott RA, Segura-Lepe MP, Shah S, Sim X, Sivapalaratnam S, Small KS, Smith AV, Smith JA, Southam L, Spector TD, Speliotes EK, Starr JM, Steinthorsdottir V, Stringham HM, Stumvoll M, Surendran P, 't Hart LM, Tansey KE, Tardif JC, Taylor KD, Teumer A, Thompson DJ, Thorsteinsdottir U, Thuesen BH, Tönjes A, Tromp G, Trompet S, Tsafantakis E, Tuomilehto J, Tybjaerg-Hansen A, Tyrer JP, Uher R, Uitterlinden AG, Ulivi S, van der Laan SW, Van Der Leij AR, van Duijn CM, van Schoor NM, van Setten J, Varbo A, Varga TV, Varma R, Edwards DRV, Vermeulen SH, Vestergaard H, Vitart V, Vogt TF, Vozzi D, Walker M, Wang F, Wang CA, Wang S, Wang Y, Wareham NJ, Warren HR, Wessel J, Willems SM, Wilson JG, Witte DR, Woods MO, Wu Y, Yaghootkar H, Yao J, Yao P, Yerges-Armstrong LM, Young R, Zeggini E, Zhan X, Zhang W, Zhao JH, Zhao W, Zhao W, Zheng H, Zhou W, Rotter JI, Boehnke M, Kathiresan S, McCarthy MI, Willer CJ, Stefansson K, Borecki IB, Liu DJ, North KE, Heard-Costa NL, Pers TH, Lindgren CM, Oxvig C, Kutalik Z, Rivadeneira F, Loos RJF, Frayling TM, Hirschhorn JN, Deloukas P, Lettre G. Rare and low-frequency coding variants alter human adult height. Nature 2017; 542:186-190. [PMID: 28146470 PMCID: PMC5302847 DOI: 10.1038/nature21039] [Show More Authors] [Citation(s) in RCA: 419] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 12/04/2016] [Indexed: 02/07/2023]
Abstract
Height is a highly heritable, classic polygenic trait with approximately 700 common associated variants identified through genome-wide association studies so far. Here, we report 83 height-associated coding variants with lower minor-allele frequencies (in the range of 0.1-4.8%) and effects of up to 2 centimetres per allele (such as those in IHH, STC2, AR and CRISPLD2), greater than ten times the average effect of common variants. In functional follow-up studies, rare height-increasing alleles of STC2 (giving an increase of 1-2 centimetres per allele) compromised proteolytic inhibition of PAPP-A and increased cleavage of IGFBP-4 in vitro, resulting in higher bioavailability of insulin-like growth factors. These 83 height-associated variants overlap genes that are mutated in monogenic growth disorders and highlight new biological candidates (such as ADAMTS3, IL11RA and NOX4) and pathways (such as proteoglycan and glycosaminoglycan synthesis) involved in growth. Our results demonstrate that sufficiently large sample sizes can uncover rare and low-frequency variants of moderate-to-large effect associated with polygenic human phenotypes, and that these variants implicate relevant genes and pathways.
Collapse
Affiliation(s)
- Eirini Marouli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands.,Department of Internal Medicine, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands
| | - Ken Sin Lo
- Montreal Heart Institute, Montreal, Quebec H1T 1C8, Canada
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Troels R Kjaer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, 8000, Denmark
| | - Rebecca S Fine
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Yingchang Lu
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37203, USA.,The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,The Genetics of Obesity and Related Metabolic Traits Program, Ichan School of Medicine at Mount Sinai, New York, New York 10069, USA
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,The Genetics of Obesity and Related Metabolic Traits Program, Ichan School of Medicine at Mount Sinai, New York, New York 10069, USA
| | - Heather M Highland
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27514, USA.,Human Genetics Center, The University of Texas School of Public Health, The University of Texas Graduate School of Biomedical Sciences at Houston, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Sina Rüeger
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | | | - Anne E Justice
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - David Lamparter
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland.,Department of Computational Biology, University of Lausanne, Lausanne, 1011, Switzerland
| | - Kathleen E Stirrups
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.,Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK
| | - Valérie Turcot
- Montreal Heart Institute, Montreal, Quebec H1T 1C8, Canada
| | - Kristin L Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, D-93051, Germany
| | - Tõnu Esko
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Tugce Karaderi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Adam E Locke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA.,McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, Missouri 63108, USA
| | - Nicholas G D Masca
- Department of Cardiovascular Sciences, Univeristy of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK.,NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Maggie C Y Ng
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA.,Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Poorva Mudgal
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Manuel A Rivas
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Nuffield Department of Clinical Medicine, Oxford OX3 7BN, UK
| | - Sailaja Vedantam
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Goncalo Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Katja K Aben
- Netherlands Comprehensive Cancer Organisation, Utrecht, 3501 DB, The Netherlands.,Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Linda S Adair
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Dewan S Alam
- Centre for Control of Chronic Diseases (CCCD), Dhaka, 1212, Bangladesh
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, D-85764, Germany
| | - Kristine H Allin
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Matthew Allison
- Department of Family Medicine &Public Health, University of California, San Diego, La Jolla, California 92093, USA
| | - Philippe Amouyel
- INSERM U1167, Lille, F-59019, France.,Institut Pasteur de Lille, U1167, Lille, F-59019, France.,Universite de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Lille, F-59019, France
| | - Emil V Appel
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Dominique Arveiler
- Department of Epidemiology and Public Health, University of Strasbourg, Strasbourg, F-67085, France.,Department of Public Health, University Hospital of Strasbourg, Strasbourg, 67081, France
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart &Lungs, University Medical Center Utrecht, Utrecht, The Netherlands.,Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands.,Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
| | - Paul L Auer
- Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201, USA
| | - Beverley Balkau
- INSERM U1018, Centre de recherche en Épidemiologie et Sante des Populations (CESP), Villejuif, France
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, 93042, Germany
| | - Lia E Bang
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, 2100, Denmark
| | - Marianne Benn
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, 2730, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Sven Bergmann
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland.,Department of Computational Biology, University of Lausanne, Lausanne, 1011, Switzerland
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Matthias Blüher
- IFB Adiposity Diseases, University of Leipzig, Leipzig, 04103, Germany.,University of Leipzig, Department of Medicine, Leipzig, 04103, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, 14558, Germany
| | - Eric Boerwinkle
- School of Public Health, Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, 93042, Germany
| | - Lori L Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jette Bork-Jensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Donald W Bowden
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA.,Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA.,Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Ivan Brandslund
- Department of Clinical Biochemistry, Lillebaelt Hospital, Vejle, 7100, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, 5000, Denmark
| | - Gerome Breen
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London &NIHR Biomedical Research Centre for Mental Health at the Maudsley, London, SE5 8AF, UK
| | | | - Linda Broer
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands
| | - Amber A Burt
- Department of Medicine, University of Washington, Seattle, Washington 98195, USA
| | - Adam S Butterworth
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge CB1 8RN, UK
| | - David J Carey
- The Sigfried and Janet Weis Center for Research, Danville, Pennsylvania 17822, USA
| | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.,NIHR Barts Cardiovascular Research Unit, Barts and The London School of Medicine &Dentistry, Queen Mary University, London EC1M 6BQ, UK
| | - John C Chambers
- Department of Cardiology, London North West Healthcare NHS Trust, Ealing Hospital, Middlesex UB1 3HW, UK.,Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK.,Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - Daniel I Chasman
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Preventive Medicine, Brigham and Women's and Harvard Medical School, Boston, Massachusetts 02215, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Rajiv Chowdhury
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | | | - Audrey Y Chu
- Division of Preventive Medicine, Brigham and Women's and Harvard Medical School, Boston, Massachusetts 02215, USA.,NHLBI Framingham Heart Study, Framingham, Massachusetts 01702, USA
| | - Massimiliano Cocca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, 34100, Italy
| | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
| | - Janie Corley
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Jordi Corominas Galbany
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Amanda J Cox
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA.,Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Gabriel Cuellar-Partida
- Diamantina Institute, University of Qeensland, Brisbane, Queensland, 4072, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge CB1 8RN, UK.,Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK.,British Heart Foundation, Cambridge Centre of Excellence, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Gail Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Paul I W de Bakker
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands
| | - Gert J de Borst
- Department of Vascular Surgery, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands
| | - Simon de Denus
- Montreal Heart Institute, Montreal, Quebec H1T 1C8, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Mark C H de Groot
- Department of Clinical Chemistry and Haematology, Division of Laboratory and Pharmacy, University Medical Center Utrecht, Utrecht, 3508 GA, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Division Pharmacoepidemiology &Clinical Pharmacology, Utrecht University, Utrecht, 3508 TB, The Netherlands
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, 17671, Greece
| | - Ellen W Demerath
- Division of Epidemiology &Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota 55454, USA
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Joe G Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Emanuele Di Angelantonio
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge CB1 8RN, UK
| | - Fotios Drenos
- Institute of Cardiovascular Science, University College London, London WC1E 6JF, UK.,MRC Integrative Epidemiology Unit, School of Social &Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Mengmeng Du
- Fred Hutchinson Cancer Research Center, Public Health Sciences Division, Seattle, Washington 98109, USA.,Memorial Sloan Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, New York 10017, USA
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Tapani Ebeling
- Department of Medicine, Oulu University Hospital, Oulu, 90029, Finland.,Research Unit of Internal Medicine, University of Oulu, Oulu, FI-90014, Finland
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee 37203, USA
| | - Patrick T Ellinor
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Medical and Population Genetics Program, Broad Institute, Cambridge, Massachusetts 02141, USA
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK.,Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, 17671, Greece
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan 48104, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, StLouis, Missouri 63108, USA
| | - Shuang Feng
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ele Ferrannini
- CNR Institute of Clinical Physiology, Pisa, Italy.,Department of Clinical &Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco M Ferrario
- Research Center on Epidemiology and Preventive Medicine, Department of Clinical and Experimental Medicine, University of Insubria, Varese, 21100, Italy
| | - Jean Ferrieres
- Toulouse University School of Medicine, Toulouse, TSA 50032 31059, France
| | - Jose C Florez
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Medical and Population Genetics Program, Broad Institute, Cambridge, Massachusetts 02141, USA.,Department of Medicine, Harvard University Medical School, Boston, Massachusetts 02115, USA
| | - Ian Ford
- University of Glasgow, Glasgow G12 8QQ, UK
| | - Myriam Fornage
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Malmö, SE-20502, Sweden.,Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA.,Department of Public Health and Clinical Medicine, Unit of Medicine, Umeå University, Umeå, 901 87, Sweden
| | - Ruth Frikke-Schmidt
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark.,Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, 2100, Denmark
| | | | - Wei Gan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Ilaria Gandin
- Department of Medical Sciences, University of Trieste, Trieste, 34137, Italy
| | - Paolo Gasparini
- Department of Medical Sciences, University of Trieste, Trieste, 34137, Italy.,Division of Experimental Genetics, Sidra Medical and Research Center, Doha, 26999, Qatar
| | - Vilmantas Giedraitis
- Geriatrics, Department of Public Health, Uppsala University, Uppsala, 751 85, Sweden
| | - Ayush Giri
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee 37203, USA
| | - Giorgia Girotto
- Department of Medical Sciences, University of Trieste, Trieste, 34137, Italy.,Division of Experimental Genetics, Sidra Medical and Research Center, Doha, 26999, Qatar
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Penny Gordon-Larsen
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina 27514, USA.,Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, D-93051, Germany.,Department of Nephrology, University Hospital Regensburg, Regensburg, 93042, Germany
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Megan L Grove
- School of Public Health, Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland.,Icelandic Heart Association, Kopavogur, 201, Iceland
| | - Stefan Gustafsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, 751 41, Sweden
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Kathleen Mullan Harris
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina 27514, USA.,Department of Sociology, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Andrew T Hattersley
- University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Caroline Hayward
- MRCHGU, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA.,Department of Public Health, University of Helsinki, Helsinki, FI-00014, Finland
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, D-93051, Germany.,Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, D-85764, Germany
| | - Kauko Heikkilä
- Department of Public Health, University of Helsinki, Helsinki, FI-00014, Finland.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland
| | - Øyvind Helgeland
- Department of Pediatrics, Haukeland University Hospital, Bergen, 5021, Norway.,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, 5020, Norway
| | - Jussi Hernesniemi
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, 33521, Finland.,Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland.,Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, 33014, Finland
| | - Alex W Hewitt
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria, 3002, Australia.,Centre for Ophthalmology and Vision Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, 6009, Australia.,Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, 7000, Australia
| | - Lynne J Hocking
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.,Musculoskeletal Research Programme, Division of Applied Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Mette Hollensted
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Oddgeir L Holmen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, NTNU, Norwegian University of Science and Technology, Trondheim, 7600, Norway
| | - G Kees Hovingh
- AMC, Department of Vascular Medicine, Amsterdam, 1105 AZ, The Netherlands
| | - Joanna M M Howson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Paul L Huang
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, 7600, Norway
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands.,Department of Neurology, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands.,Department of Radiology, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, 751 41, Sweden.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jan-Håkan Jansson
- Department of Public Health &Clinical Medicine, Umeå University, Umeå, SE-90185, Sweden.,Research Unit Skellefteå, Skellefteå, SE-93141, Sweden
| | - Gail P Jarvik
- Department of Medicine, University of Washington, Seattle, Washington 98195, USA.,Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA
| | - Gorm B Jensen
- The Copenhagen City Heart Study, Frederiksberg Hospital, Frederiksberg, 2000, Denmark
| | - Min A Jhun
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Yucheng Jia
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Xuejuan Jiang
- Department of Preventive Medicine, Keck School of Medicine of the University of California, Los Angeles, California 90089, USA.,USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California 90089, USA
| | - Stefan Johansson
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, 5020, Norway.,Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5021, Norway
| | - Marit E Jørgensen
- National Institute of Public Health, University of Southern Denmark, Copenhagen, 1353, Denmark.,Steno Diabetes Center, Gentofte, 2820, Denmark
| | - Torben Jørgensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark.,Aalborg University, Aalborg, DK-9000, Denmark.,Research Center for Prevention and Health, Capital Region of Denmark, Glostrup, DK-2600, Denmark
| | - Pekka Jousilahti
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, 2333, The Netherlands.,The Interuniversity Cardiology Institute of the Netherlands, Utrecht, 2333, The Netherlands
| | - Bratati Kahali
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA.,Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - René S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, 3584 CG, The Netherlands
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, 33014, Finland
| | - Pia R Kamstrup
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, 2730, Denmark
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, Helsinki, FI-00014, Finland.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland.,National Institute for Health and Welfare, Helsinki, FI-00271, Finland
| | | | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK.,Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | - Frank Kee
- UKCRC Centre of Excellence for Public Health Research, Queens University Belfast, Belfast BT12 6BJ, UK
| | - Renske Keeman
- Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Amsterdam, 1066 CX, The Netherlands
| | | | - Hidetoshi Kitajima
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | | | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology and Endodontology, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland
| | - Jukka Kontto
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
| | - Jaspal S Kooner
- Department of Cardiology, London North West Healthcare NHS Trust, Ealing Hospital, Middlesex UB1 3HW, UK.,Imperial College Healthcare NHS Trust, London W12 0HS, UK.,National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Peter Kovacs
- IFB Adiposity Diseases, University of Leipzig, Leipzig, 04103, Germany
| | - Jennifer Kriebel
- German Center for Diabetes Research, München-Neuherberg, 85764, Germany.,Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, D-85764, Germany.,Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, D-85764, Germany
| | - Helena Kuivaniemi
- The Sigfried and Janet Weis Center for Research, Danville, Pennsylvania 17822, USA.,Department of Psychiatry, and Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Western Cape, 7505, South Africa
| | - Sébastien Küry
- CHU Nantes, Service de Génétique Médicale, Nantes, 44093, France
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Martina La Bianca
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, 34137, Italy
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Timo A Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland.,Institute of Biomedicine &Physiology, University of Eastern Finland, Kuopio, 70210, Finland
| | - Ethan M Lange
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Leslie A Lange
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Carl D Langefeld
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle, Washington 98195, USA.,Group Health Research Institute, Seattle, Washington 98101, USA.,Department of Health Services, University of Washington, Seattle, Washington 98101, USA
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan.,School of Medicine, National Yang-Ming University, Taipei 112, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland.,Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, 33014, Finland
| | - Cora E Lewis
- Division of Preventive Medicine University of Alabama at Birmingham, Birmingham, Alabama 35205, USA
| | - Huaixing Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China, Shanghai, 200031, China
| | - Jin Li
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California 94304, USA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Honghuang Lin
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Li-An Lin
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China, Shanghai, 200031, China
| | - Lars Lind
- Uppsala University, Uppsala, 75185, Sweden
| | - Jaana Lindström
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
| | - Allan Linneberg
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark.,Research Center for Prevention and Health, Capital Region of Denmark, Glostrup, DK-2600, Denmark.,Department of Experimental Medicine, Rigshospitalet, Copenhagen, DK-2200, Denmark
| | - Yeheng Liu
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Yongmei Liu
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Artitaya Lophatananon
- Division of Health Sciences, Warwick Medical School, Warwick University, Coventry CV4 7AL, UK
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Steven A Lubitz
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Medical and Population Genetics Program, Broad Institute, Cambridge, Massachusetts 02141, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland.,Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, 33014, Finland
| | - David A Mackey
- Centre for Ophthalmology and Vision Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Pamela A F Madden
- Department of Psychiatry, Washington University, Saint Louis, Missouri 63110, USA
| | - Alisa K Manning
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Medical and Population Genetics Program, Broad Institute, Cambridge, Massachusetts 02141, USA.,Department of Medicine, Harvard University Medical School, Boston, Massachusetts 02115, USA
| | - Satu Männistö
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
| | - Gaëlle Marenne
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Jonathan Marten
- MRCHGU, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Angela L Mazul
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Karina Meidtner
- German Center for Diabetes Research, München-Neuherberg, 85764, Germany.,Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, 14558, Germany
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Paul Mitchell
- Westmead Millennium Institute of Medical Research, Centre for Vision Research and Department of Ophthalmology, University of Sydney, Sydney, New South Wales, 2022, Australia
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands.,Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Anna Morgan
- Department of Medical Sciences, University of Trieste, Trieste, 34137, Italy
| | - Andrew D Morris
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh EH8 9AG, 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
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, D-85764, Germany.,Department of Medicine I, Ludwig-Maximilians-Universität, Munich, 81377, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, 80802, Germany
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.,NIHR Barts Cardiovascular Research Unit, Barts and The London School of Medicine &Dentistry, Queen Mary University, London EC1M 6BQ, UK
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, Maryland 20892, USA
| | - Matthias Nauck
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, 17475, Germany.,Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, Univeristy of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK.,NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Matt Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK.,Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | - Sune F Nielsen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, 2730, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, 33521, Finland
| | - Pål R Njølstad
- Department of Pediatrics, Haukeland University Hospital, Bergen, 5021, Norway.,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, 5020, Norway
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, 2730, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jeffrey R O'Connel
- Program in Personalized Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Heikki Oksa
- Department of Medicine, Tampere University Hospital, Tampere, 33521, Finland
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, UCLA, Los Angeles, California 90095, USA
| | - Roel A Ophoff
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, 3584 CG, The Netherlands.,Center for Neurobehavioral Genetics, UCLA, Los Angeles, California 90095, USA
| | - Katharine R Owen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK.,Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | | | | | - Colin N A Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Medical Research Institute, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands.,Laboratory of Experimental Cardiology, Division Heart &Lungs, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands
| | - Aniruddh P Patel
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA.,Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Alison Pattie
- Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Peggy L Peissig
- Marshfield Clinic Research Foundation, Marshfield, Wisconsin 54449, USA
| | - Gina M Peloso
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Medical and Population Genetics Program, Broad Institute, Cambridge, Massachusetts 02141, USA
| | - Craig E Pennell
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland.,University of Helsinki, Institute for Molecular Medicine (FIMM) and Diabetes and Obesity Research Program, Helsinki, FI00014, Finland.,University of Tartu, Estonian Genome Center, Tartu, Estonia, Tartu, 51010, Estonia
| | - James A Perry
- Program in Personalized Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Thomas N Person
- Marshfield Clinic Research Foundation, Marshfield, Wisconsin 54449, USA
| | - Ailith Pirie
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Ozren Polasek
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh EH8 9AG, UK.,School of Medicine, University of Split, Split, 21000, Croatia
| | - Danielle Posthuma
- Center for Neurogenomics and Cognitive Research, Department Complex Trait Genetics, VU University, Amsterdam, 1081 HV, The Netherlands.,Neuroscience Campus Amsterdam, Department Clinical Genetics, VU Medical Center, Amsterdam, 1081 HV, The Netherlands
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, 20521, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20520, Finland
| | - Asif Rasheed
- Centre for Non-Communicable Diseases, Karachi, Pakistan
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland.,Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, 70210, Finland
| | - Dermot F Reilly
- MRL, Merck &Co., Inc., Genetics and Pharmacogenomics, Boston, Massachusetts 02115, USA
| | - Alex P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.,Department of Epidemiology, University of Washington, Seattle, Washington 98195, USA
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Malmö, SE-20502, Sweden.,Department of Biobank Research, Umeå University, Umeå, SE-90187, Sweden
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's and Harvard Medical School, Boston, Massachusetts 02215, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - John D Rioux
- Montreal Heart Institute, Montreal, Quebec H1T 1C8, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Neil Robertson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK
| | - Antonietta Robino
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, 34137, Italy
| | - Olov Rolandsson
- Department of Public Health &Clinical Medicine, Umeå University, Umeå, SE-90185, Sweden.,Department of Public Health and Clinical Medicine, Unit of Family Medicine, Umeå University, Umeå, 90185, Sweden
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Katherine S Ruth
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Danish Saleheen
- Centre for Non-Communicable Diseases, Karachi, Pakistan.,Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, Univeristy of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK.,NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Kevin Sandow
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Yadav Sapkota
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | | | - Marjanka K Schmidt
- Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Amsterdam, 1066 CX, The Netherlands
| | - Pamela J Schreiner
- Division of Epidemiology &Community Health University of Minnesota, Minneapolis, Minnesota 55454, USA
| | - Matthias B Schulze
- German Center for Diabetes Research, München-Neuherberg, 85764, Germany.,Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, 14558, Germany
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Marcelo P Segura-Lepe
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Svati Shah
- Duke University, Durham, North Carolina 27703, USA
| | - Xueling Sim
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA.,Saw Swee Hock School of Public Health, National University of Singapore, National University Health System, Singapore, Singapore
| | - Suthesh Sivapalaratnam
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Departement of Haematology, University of Cambridge, Cambridge CB2 OPT, UK.,Department of Vascular Medicine, AMC, Amsterdam, 1105 AZ, The Netherlands
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, Kingís College London, London SE1 7EH, UK
| | - Albert Vernon Smith
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland.,Icelandic Heart Association, Kopavogur, 201, Iceland
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Lorraine Southam
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, Kingís College London, London SE1 7EH, UK
| | - Elizabeth K Speliotes
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA.,Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | | | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Michael Stumvoll
- IFB Adiposity Diseases, University of Leipzig, Leipzig, 04103, Germany.,University of Leipzig, Department of Medicine, Leipzig, 04103, Germany
| | - Praveen Surendran
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Leen M 't Hart
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, 1007MB, The Netherlands.,Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, 2333ZC, The Netherlands.,Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, 2333ZC, The Netherlands
| | - Katherine E Tansey
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4EP, UK.,MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec H1T 1C8, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Deborah J Thompson
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen inc., Reykjavik, 101, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Betina H Thuesen
- Research Center for Prevention and Health, Capital Region of Denmark, Glostrup, DK-2600, Denmark
| | - Anke Tönjes
- Center for Pediatric Research, Department for Women's and Child Health, University of Leipzig, Leipzig, 04103, Germany
| | - Gerard Tromp
- The Sigfried and Janet Weis Center for Research, Danville, Pennsylvania 17822, USA.,Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Western Cape, 7505, South Africa
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, 2333, The Netherlands.,Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, 2333, The Netherlands
| | | | - Jaakko Tuomilehto
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland.,Centre for Vascular Prevention, Danube-University Krems, Krems, 3500, Austria.,Dasman Diabetes Institute, Dasman, 15462, Kuwait.,Diabetes Research Group, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - 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
| | - Jonathan P Tyrer
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Rudolf Uher
- Department of Psychiatry, Dalhousie University, Halifax B3H 4R2, Canada
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands.,Department of Internal Medicine, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands
| | - Sheila Ulivi
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, 34137, Italy
| | - Sander W van der Laan
- Laboratory of Experimental Cardiology, Division Heart &Lungs, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands
| | - Andries R Van Der Leij
- University of Amsterdam, Department of Brain &Cognition, Amsterdam, 1018 WS, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, 1007MB, The Netherlands
| | - Jessica van Setten
- Department of Cardiology, Division Heart &Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anette Varbo
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, 2730, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Tibor V Varga
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Malmö, SE-20502, Sweden
| | - Rohit Varma
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California 90089, USA
| | - Digna R Velez Edwards
- Department of Obstetrics and Gynecology, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee 37203, USA
| | - Sita H Vermeulen
- Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Henrik Vestergaard
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Veronique Vitart
- MRCHGU, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Thomas F Vogt
- MRL, Merck &Co., Inc., Cardiometabolic Disease, Kenilworth, New Jersey 07033, USA
| | - Diego Vozzi
- Division of Experimental Genetics, Sidra Medical and Research Center, Doha, 26999, Qatar
| | - Mark Walker
- Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle NE2 4HH, UK
| | - Feijie Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China, Shanghai, 200031, China
| | - Carol A Wang
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Shuai Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Yiqin Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China, Shanghai, 200031, China
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.,NIHR Barts Cardiovascular Research Unit, Barts and The London School of Medicine &Dentistry, Queen Mary University, London EC1M 6BQ, UK
| | - Jennifer Wessel
- Departments of Epidemiology &Medicine, Diabetes Translational Research Center, Fairbanks School of Public Health &School of Medicine, Indiana University, Indiana 46202, USA
| | - Sara M Willems
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | - Daniel R Witte
- Danish Diabetes Academy, Odense, 5000, Denmark.,Department of Public Health, Aarhus University, Aarhus, 8000, Denmark
| | - Michael O Woods
- Memorial University, Faculty of Medicine, Discipline of Genetics, St. John's, Newfoundland A1B 3V6, Canada
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Pang Yao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China, Shanghai, 200031, China
| | - Laura M Yerges-Armstrong
- Program in Personalized Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.,GlaxoSmithKlein, King of Prussia, Pennsylvania 19406, USA
| | - Robin Young
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,University of Glasgow, Glasgow G12 8QQ, UK
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Xiaowei Zhan
- Department of Clinical Sciences, Quantitative Biomedical Research Center, Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Weihua Zhang
- Department of Cardiology, London North West Healthcare NHS Trust, Ealing Hospital, Middlesex UB1 3HW, UK.,Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Wei Zhao
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - He Zheng
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China, Shanghai, 200031, China
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | | | | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Sekar Kathiresan
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA.,Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK.,Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | - Cristen J Willer
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Kari Stefansson
- deCODE Genetics/Amgen inc., Reykjavik, 101, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, StLouis, Missouri 63108, USA
| | - Dajiang J Liu
- Department of Public Health Sciences, Institute for Personalized Medicine, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Kari E North
- Department of Epidemiology and Carolina Center of Genome Sciences, Chapel Hill, North Carolina 27514, USA
| | - Nancy L Heard-Costa
- NHLBI Framingham Heart Study, Framingham, Massachusetts 01702, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Tune H Pers
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark.,Department of Epidemiology Research, Statens Serum Institut, Copenhagen, 2200, Denmark
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,Li Ka Shing Centre for Health Information and Discovery, The Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, 8000, Denmark
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands.,Department of Internal Medicine, Erasmus Medical Center, Rotterdam, 3015 GE, The Netherlands
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.,The Genetics of Obesity and Related Metabolic Traits Program, Ichan School of Medicine at Mount Sinai, New York, New York 10069, USA.,The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, New York 10069, USA
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Joel N Hirschhorn
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Departments of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.,Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, Quebec H1T 1C8, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| |
Collapse
|
977
|
A molecular census of arcuate hypothalamus and median eminence cell types. Nat Neurosci 2017; 20:484-496. [PMID: 28166221 PMCID: PMC5323293 DOI: 10.1038/nn.4495] [Citation(s) in RCA: 567] [Impact Index Per Article: 70.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 01/04/2017] [Indexed: 02/07/2023]
Abstract
The hypothalamic arcuate-median eminence complex (Arc-ME) controls energy balance, fertility, and growth through molecularly distinct cell types, many of which remain unknown. To catalog cell types in an unbiased way, we profiled gene expression in 20,921 individual cells in and around the adult mouse Arc-ME using Drop-seq. We identify 50 transcriptionally distinct Arc-ME cell populations, including a rare tanycyte population at the Arc-ME diffusion barrier, a novel leptin-sensing neuronal population, multiple AgRP and POMC subtypes, and an orexigenic somatostatin neuronal population. We extended Drop-seq to detect dynamic expression changes across relevant physiological perturbations, revealing cell type-specific responses to energy status, including distinctly responsive subtypes of AgRP and POMC neurons. Finally, integrating our data with human GWAS data implicates two previously unknown neuronal subtypes in the genetic control of obesity. This resource will accelerate biological discovery by providing insights into molecular and cell type diversity from which function can be inferred.
Collapse
|
978
|
Gkourogianni A, Andrew M, Tyzinski L, Crocker M, Douglas J, Dunbar N, Fairchild J, Funari MFA, Heath KE, Jorge AAL, Kurtzman T, LaFranchi S, Lalani S, Lebl J, Lin Y, Los E, Newbern D, Nowak C, Olson M, Popovic J, Průhová Š, Elblova L, Quintos JB, Segerlund E, Sentchordi L, Shinawi M, Stattin EL, Swartz J, del Angel AG, Cuéllar SD, Hosono H, Sanchez-Lara PA, Hwa V, Baron J, Nilsson O, Dauber A. Clinical Characterization of Patients With Autosomal Dominant Short Stature due to Aggrecan Mutations. J Clin Endocrinol Metab 2017; 102:460-469. [PMID: 27870580 PMCID: PMC5413162 DOI: 10.1210/jc.2016-3313] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/18/2016] [Indexed: 12/22/2022]
Abstract
CONTEXT Heterozygous mutations in the aggrecan gene (ACAN) cause autosomal dominant short stature with accelerated skeletal maturation. OBJECTIVE We sought to characterize the phenotypic spectrum and response to growth-promoting therapies. PATIENTS AND METHODS One hundred three individuals (57 females, 46 males) from 20 families with autosomal dominant short stature and heterozygous ACAN mutations were identified and confirmed using whole-exome sequencing, targeted next-generation sequencing, and/or Sanger sequencing. Clinical information was collected from the medical records. RESULTS Identified ACAN variants showed perfect cosegregation with phenotype. Adult individuals had mildly disproportionate short stature [median height, -2.8 standard deviation score (SDS); range, -5.9 to -0.9] and a history of early growth cessation. The condition was frequently associated with early-onset osteoarthritis (12 families) and intervertebral disc disease (9 families). No apparent genotype-phenotype correlation was found between the type of ACAN mutation and the presence of joint complaints. Childhood height was less affected (median height, -2.0 SDS; range, -4.2 to -0.6). Most children with ACAN mutations had advanced bone age (bone age - chronologic age; median, +1.3 years; range, +0.0 to +3.7 years). Nineteen individuals had received growth hormone therapy with some evidence of increased growth velocity. CONCLUSIONS Heterozygous ACAN mutations result in a phenotypic spectrum ranging from mild and proportionate short stature to a mild skeletal dysplasia with disproportionate short stature and brachydactyly. Many affected individuals developed early-onset osteoarthritis and degenerative disc disease, suggesting dysfunction of the articular cartilage and intervertebral disc cartilage. Additional studies are needed to determine the optimal treatment strategy for these patients.
Collapse
Affiliation(s)
- Alexandra Gkourogianni
- Division of Pediatric Endocrinology, Department of Women’s and Children’s Health, Karolinska Institutet and Karolinska University Hospital, Stockholm SE-171 76, Sweden
| | - Melissa Andrew
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 70941
| | - Leah Tyzinski
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 70941
| | | | - Jessica Douglas
- Genetics, Boston Children’s Hospital, Boston, Massachusetts 02115
| | - Nancy Dunbar
- Division of Pediatric Endocrinology, Connecticut Children’s Medical Center, Hartford, Connecticut 06106
| | - Jan Fairchild
- Department of Endocrinology and Diabetes, Women's and Children's Hospital, North Adelaide, South Australia 5006, Australia
| | - Mariana F. A. Funari
- Unidade de Endocrinologia do Desenvolvimento (LIM/42), Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05508-020, Brazil
| | - Karen E. Heath
- Institute of Medical and Molecular Genetics (INGEMM) and Skeletal Dysplasia Multidisciplinary Unit, Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, and CIBERER, ISCIII, Madrid 20849, Spain
| | - Alexander A. L. Jorge
- Unidade de Endocrinologia do Desenvolvimento (LIM/42), Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05508-020, Brazil
| | | | - Stephen LaFranchi
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97239
| | | | - Jan Lebl
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital in Motol, Prague 11636, Czech Republic
| | - Yuezhen Lin
- Pediatric Endocrinology and Metabolism, Baylor College of Medicine, Houston, Texas 77030
| | - Evan Los
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97239
| | - Dorothee Newbern
- Division of Endocrinology, Phoenix Children’s Hospital, Phoenix, Arizona 85016
| | - Catherine Nowak
- Genetics, Boston Children’s Hospital, Boston, Massachusetts 02115
| | - Micah Olson
- Division of Endocrinology, Phoenix Children’s Hospital, Phoenix, Arizona 85016
| | - Jadranka Popovic
- Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15237
| | - Štěpánka Průhová
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital in Motol, Prague 11636, Czech Republic
| | - Lenka Elblova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital in Motol, Prague 11636, Czech Republic
| | | | - Emma Segerlund
- Division of Pediatric Endocrinology, Department of Women’s and Children’s Health, Karolinska Institutet and Karolinska University Hospital, Stockholm SE-171 76, Sweden
- Sunderby Hospital, Sunderby 95442, Sweden
| | - Lucia Sentchordi
- Institute of Medical and Molecular Genetics (INGEMM) and Skeletal Dysplasia Multidisciplinary Unit, Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, and CIBERER, ISCIII, Madrid 20849, Spain
- Department of Pediatrics, Hospital Universitario Infanta Sofia, Madrid 28703, Spain
| | - Marwan Shinawi
- Division of Genetics, Washington University, St. Louis, Missouri 63130
| | - Eva-Lena Stattin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala 75236, Sweden
| | | | - Ariadna González del Angel
- Laboratorio de Biología Molecular, Departamento de Genética Humana, Instituto Nacional de Pediatría, Insurgentes-Cuicuilco, Coyoacán 04530, México
| | - Sinhué Diaz Cuéllar
- Laboratorio de Biología Molecular, Departamento de Genética Humana, Instituto Nacional de Pediatría, Insurgentes-Cuicuilco, Coyoacán 04530, México
| | - Hidekazu Hosono
- Cottage Children’s Medical Center, Santa Barbara, California 93111
| | - Pedro A. Sanchez-Lara
- Center for Personalized Medicine, Children’s Hospital of Los Angeles, Los Angeles, California 90027
| | - Vivian Hwa
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 70941
| | - Jeffrey Baron
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; and
| | - Ola Nilsson
- Division of Pediatric Endocrinology, Department of Women’s and Children’s Health, Karolinska Institutet and Karolinska University Hospital, Stockholm SE-171 76, Sweden
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; and
- Department of Medical Sciences, Örebro University and University Hospital, Örebro 70185, Sweden
| | - Andrew Dauber
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 70941
| |
Collapse
|
979
|
Böckerman P, Viinikainen J, Vainiomäki J, Hintsanen M, Pitkänen N, Lehtimäki T, Pehkonen J, Rovio S, Raitakari O. Stature and long-term labor market outcomes: Evidence using Mendelian randomization. ECONOMICS AND HUMAN BIOLOGY 2017; 24:18-29. [PMID: 27846416 DOI: 10.1016/j.ehb.2016.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/21/2016] [Accepted: 10/27/2016] [Indexed: 06/06/2023]
Abstract
We use the Young Finns Study (N=∼2000) on the measured height linked to register-based long-term labor market outcomes. The data contain six age cohorts (ages 3, 6, 9, 12, 15 and 18, in 1980) with the average age of 31.7, in 2001, and with the female share of 54.7. We find that taller people earn higher earnings according to the ordinary least squares (OLS) estimation. The OLS models show that 10cm of extra height is associated with 13% higher earnings. We use Mendelian randomization, with the genetic score as an instrumental variable (IV) for height to account for potential confounders that are related to socioeconomic background, early life conditions and parental investments, which are otherwise very difficult to fully account for when using covariates in observational studies. The IV point estimate is much lower and not statistically significant, suggesting that the OLS estimation provides an upward biased estimate for the height premium. Our results show the potential value of using genetic information to gain new insights into the determinants of long-term labor market success.
Collapse
Affiliation(s)
- Petri Böckerman
- Turku School of Economics, Labour Institute for Economic Research, Helsinki, Finland.
| | - Jutta Viinikainen
- Jyväskylä University School of Business and Economics, Jyväskylä, Finland
| | | | | | - Niina Pitkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Finland
| | - Jaakko Pehkonen
- Jyväskylä University School of Business and Economics, Jyväskylä, Finland
| | - Suvi Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
980
|
Abstract
PURPOSE OF REVIEW As the incidence of type 1 diabetes (T1DM) continues to rise, complications including impairment of childhood growth remain a major concern. This review provides an overview of alterations in growth patterns before and after the onset of T1DM. RECENT FINDINGS Recent advances in this field include several prospective investigations of height and weight trajectories in children leading up to the development of islet autoimmunity and T1DM as well as evaluations of larger cohorts of T1DM patients to better assess predictors of altered growth. In addition, genetic and metabolic investigations have improved our understanding of the more rare severe growth impairment of Mauriac Syndrome. SUMMARY Despite advances in medical care of children with T1DM, growth remains suboptimal in this population and likely reflects ongoing metabolic derangement linked with classic microvascular diabetic complications.
Collapse
Affiliation(s)
- Deborah M Mitchell
- Endocrine Unit and Pediatric Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
981
|
Cesarini D, Visscher PM. Genetics and educational attainment. NPJ SCIENCE OF LEARNING 2017; 2:4. [PMID: 30631451 PMCID: PMC6220209 DOI: 10.1038/s41539-017-0005-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 05/25/2023]
Abstract
We explore how advances in our understanding of the genetics of complex traits such as educational attainment could constructively be leveraged to advance research on education and learning. We discuss concepts and misconceptions about genetic findings with regard to causes, consequences, and policy. Our main thesis is that educational attainment as a measure that varies between individuals in a population can be subject to exactly the same experimental biological designs as other outcomes, for example, those studied in epidemiology and medical sciences, and the same caveats about interpretation and implication apply.
Collapse
Affiliation(s)
- David Cesarini
- Department of Economics, New York University, New York, NY 10012 United States
| | - Peter M. Visscher
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072 Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia
| |
Collapse
|
982
|
Pasaniuc B, Price AL. Dissecting the genetics of complex traits using summary association statistics. Nat Rev Genet 2017; 18:117-127. [PMID: 27840428 PMCID: PMC5449190 DOI: 10.1038/nrg.2016.142] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the past decade, genome-wide association studies (GWAS) have been used to successfully identify tens of thousands of genetic variants associated with complex traits and diseases. These studies have produced extensive repositories of genetic variation and trait measurements across large numbers of individuals, providing tremendous opportunities for further analyses. However, privacy concerns and other logistical considerations often limit access to individual-level genetic data, motivating the development of methods that analyse summary association statistics. Here, we review recent progress on statistical methods that leverage summary association data to gain insights into the genetic basis of complex traits and diseases.
Collapse
Affiliation(s)
- Bogdan Pasaniuc
- Departments of Human Genetics, and Pathology and Laboratory Medicine, University of California, Los Angeles, California 90095, USA
| | - Alkes L Price
- Departments of Epidemiology and Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
983
|
Pes GM, Tognotti E, Poulain M, Chambre D, Dore MP. Why were Sardinians the shortest Europeans? A journey through genes, infections, nutrition, and sex. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2017; 163:3-13. [PMID: 28138956 DOI: 10.1002/ajpa.23177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/28/2016] [Accepted: 01/04/2017] [Indexed: 01/12/2023]
Abstract
Since ancient times the Mediterranean island of Sardinia has been known for harboring a population with an average body height shorter than almost every other ethnic group in Europe. After over a century of investigations, the cause(s) at the origin of this uniqueness are not yet clear. The shorter stature of Sardinians appears to have been documented since prehistoric times, as revealed by the analysis of skeletal remains discovered in archaeological sites on the island. Recently, a number of genetic, hormonal, environmental, infective and nutritional factors have been put forward to explain this unique anthropometric feature, which persisted for a long time, even when environmental and living conditions improved around 1960. Although some of the putative factors are supported by sound empirical evidence, weaker support is available for others. The recent advent of whole genome analysis techniques shed new light on specific variants at the origin of this short stature. However, the marked geographical variability of stature across time and space within the island, and the well-known presence of pockets of short height in the population of the southern districts, are still puzzling findings that have attracted the interest of anthropologists and geneticists. The purpose of this review is to focus on the state-of-the-art research on stature, as well as the factors that made Sardinians the shortest among Europeans.
Collapse
Affiliation(s)
- Giovanni Mario Pes
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, 07100, Italy
| | - Eugenia Tognotti
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, 07100, Italy
| | - Michel Poulain
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, 07100, Italy
| | - Dany Chambre
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, 07100, Italy
| | - Maria Pina Dore
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, 07100, Italy
| |
Collapse
|
984
|
Genome-wide association study of prostate-specific antigen levels identifies novel loci independent of prostate cancer. Nat Commun 2017; 8:14248. [PMID: 28139693 PMCID: PMC5290311 DOI: 10.1038/ncomms14248] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/12/2016] [Indexed: 12/22/2022] Open
Abstract
Prostate-specific antigen (PSA) levels have been used for detection and surveillance of prostate cancer (PCa). However, factors other than PCa—such as genetics—can impact PSA. Here we present findings from a genome-wide association study (GWAS) of PSA in 28,503 Kaiser Permanente whites and 17,428 men from replication cohorts. We detect 40 genome-wide significant (P<5 × 10−8) single-nucleotide polymorphisms (SNPs): 19 novel, 15 previously identified for PSA (14 of which were also PCa-associated), and 6 previously identified for PCa only. Further analysis incorporating PCa cases suggests that at least half of the 40 SNPs are PSA-associated independent of PCa. The 40 SNPs explain 9.5% of PSA variation in non-Hispanic whites, and the remaining GWAS SNPs explain an additional 31.7%; this percentage is higher in younger men, supporting the genetic basis of PSA levels. These findings provide important information about genetic markers for PSA that may improve PCa screening, thereby reducing over-diagnosis and over-treatment. Prostate-specific antigen is used as a biomarker of prostate cancer, but levels can be affected by other factors not related to cancer. Here, the authors find genes associated with prostate specific antigen levels in healthy men, which could be used to reduce over-diagnosis and over-treatment.
Collapse
|
985
|
From exomes to genomes: challenges and solutions in population-based genetic association studies. Eur J Hum Genet 2017; 25:395-396. [PMID: 28120836 DOI: 10.1038/ejhg.2016.206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
986
|
Laakso M, Kuusisto J, Stančáková A, Kuulasmaa T, Pajukanta P, Lusis AJ, Collins FS, Mohlke KL, Boehnke M. The Metabolic Syndrome in Men study: a resource for studies of metabolic and cardiovascular diseases. J Lipid Res 2017; 58:481-493. [PMID: 28119442 DOI: 10.1194/jlr.o072629] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/15/2017] [Indexed: 12/30/2022] Open
Abstract
The Metabolic Syndrome in Men (METSIM) study is a population-based study including 10,197 Finnish men examined in 2005-2010. The aim of the study is to investigate nongenetic and genetic factors associated with the risk of T2D and CVD, and with cardiovascular risk factors. The protocol includes a detailed phenotyping of the participants, an oral glucose tolerance test, fasting laboratory measurements including proton NMR measurements, mass spectometry metabolomics, adipose tissue biopsies from 1,400 participants, and a stool sample. In our ongoing follow-up study, we have, to date, reexamined 6,496 participants. Extensive genotyping and exome sequencing have been performed for essentially all METSIM participants, and >2,000 METSIM participants have been whole-genome sequenced. We have identified several nongenetic markers associated with the development of diabetes and cardiovascular events, and participated in several genetic association studies to identify gene variants associated with diabetes, hyperglycemia, and cardiovascular risk factors. The generation of a phenotype and genotype resource in the METSIM study allows us to proceed toward a "systems genetics" approach, which includes statistical methods to quantitate and integrate intermediate phenotypes, such as transcript, protein, or metabolite levels, to provide a global view of the molecular architecture of complex traits.
Collapse
Affiliation(s)
- Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland .,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Päivi Pajukanta
- Departments of Human Genetics David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Aldons J Lusis
- Departments of Human Genetics David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
987
|
Wells JC. Worldwide variability in growth and its association with health: Incorporating body composition, developmental plasticity, and intergenerational effects. Am J Hum Biol 2017; 29. [DOI: 10.1002/ajhb.22954] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/24/2016] [Accepted: 12/10/2016] [Indexed: 12/11/2022] Open
Affiliation(s)
- Jonathan C.K. Wells
- UCL Great Ormond Street Institute of Child Health; 30 Guilford Street London WC1N 1EH United Kingdom
| |
Collapse
|
988
|
de Vries PS, Sabater-Lleal M, Chasman DI, Trompet S, Ahluwalia TS, Teumer A, Kleber ME, Chen MH, Wang JJ, Attia JR, Marioni RE, Steri M, Weng LC, Pool R, Grossmann V, Brody JA, Venturini C, Tanaka T, Rose LM, Oldmeadow C, Mazur J, Basu S, Frånberg M, Yang Q, Ligthart S, Hottenga JJ, Rumley A, Mulas A, de Craen AJM, Grotevendt A, Taylor KD, Delgado GE, Kifley A, Lopez LM, Berentzen TL, Mangino M, Bandinelli S, Morrison AC, Hamsten A, Tofler G, de Maat MPM, Draisma HHM, Lowe GD, Zoledziewska M, Sattar N, Lackner KJ, Völker U, McKnight B, Huang J, Holliday EG, McEvoy MA, Starr JM, Hysi PG, Hernandez DG, Guan W, Rivadeneira F, McArdle WL, Slagboom PE, Zeller T, Psaty BM, Uitterlinden AG, de Geus EJC, Stott DJ, Binder H, Hofman A, Franco OH, Rotter JI, Ferrucci L, Spector TD, Deary IJ, März W, Greinacher A, Wild PS, Cucca F, Boomsma DI, Watkins H, Tang W, Ridker PM, Jukema JW, Scott RJ, Mitchell P, Hansen T, O'Donnell CJ, Smith NL, Strachan DP, Dehghan A. Comparison of HapMap and 1000 Genomes Reference Panels in a Large-Scale Genome-Wide Association Study. PLoS One 2017; 12:e0167742. [PMID: 28107422 PMCID: PMC5249120 DOI: 10.1371/journal.pone.0167742] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/19/2016] [Indexed: 12/21/2022] Open
Abstract
An increasing number of genome-wide association (GWA) studies are now using the higher resolution 1000 Genomes Project reference panel (1000G) for imputation, with the expectation that 1000G imputation will lead to the discovery of additional associated loci when compared to HapMap imputation. In order to assess the improvement of 1000G over HapMap imputation in identifying associated loci, we compared the results of GWA studies of circulating fibrinogen based on the two reference panels. Using both HapMap and 1000G imputation we performed a meta-analysis of 22 studies comprising the same 91,953 individuals. We identified six additional signals using 1000G imputation, while 29 loci were associated using both HapMap and 1000G imputation. One locus identified using HapMap imputation was not significant using 1000G imputation. The genome-wide significance threshold of 5×10-8 is based on the number of independent statistical tests using HapMap imputation, and 1000G imputation may lead to further independent tests that should be corrected for. When using a stricter Bonferroni correction for the 1000G GWA study (P-value < 2.5×10-8), the number of loci significant only using HapMap imputation increased to 4 while the number of loci significant only using 1000G decreased to 5. In conclusion, 1000G imputation enabled the identification of 20% more loci than HapMap imputation, although the advantage of 1000G imputation became less clear when a stricter Bonferroni correction was used. More generally, our results provide insights that are applicable to the implementation of other dense reference panels that are under development.
Collapse
Affiliation(s)
- Paul S. de Vries
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Maria Sabater-Lleal
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Tarunveer S. Ahluwalia
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Novo Nordisk Foundation Center For Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marcus E. Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ming-Huei Chen
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States of America
- Framingham Heart Study, Population Sciences Branch, Division of Intramural Research National Heart Lung and Blood Institute, National Institutes of Health, Framingham, MA, United States of America
| | - Jie Jin Wang
- Centre for Vision Research, Department of Ophthalmology, and Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - John R. Attia
- Public Health Stream, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Riccardo E. Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Lu-Chen Weng
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, United States of America
| | - Rene Pool
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, the Netherlands
- EMGO+ institute, VU University & VU medical center, Amsterdam, the Netherlands
| | - Vera Grossmann
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jennifer A. Brody
- Department of Medicine, University of Washington, Seattle WA, United States of America
| | - Cristina Venturini
- Division of Infection and Immunology, UCL, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Christopher Oldmeadow
- Public Health Stream, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Johanna Mazur
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Saonli Basu
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, United States of America
| | - Mattias Frånberg
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Numerical Analysis and Computer Science, Stockholm University, Stockholm, Sweden
| | - Qiong Yang
- Framingham Heart Study, Population Sciences Branch, Division of Intramural Research National Heart Lung and Blood Institute, National Institutes of Health, Framingham, MA, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | - Symen Ligthart
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Jouke J. Hottenga
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, the Netherlands
| | - Ann Rumley
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne Grotevendt
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Kent D. Taylor
- 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, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Graciela E. Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Annette Kifley
- Centre for Vision Research, Department of Ophthalmology, and Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Lorna M. Lopez
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Royal College of Surgeons in Ireland, Department of Psychiatry, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
- University College Dublin, UCD Conway Institute, Centre for Proteome Research, UCD, Belfield, Dublin, Ireland
| | - Tina L. Berentzen
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
- NIHR Biomedical Research Centre at Guy’s and St. Thomas’ Foundation Trust, London, United Kingdom
| | | | | | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Geoffrey Tofler
- Royal North Shore Hospital, Sydney University, Sydney, Australia
| | | | - Harmen H. M. Draisma
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, the Netherlands
- Neuroscience Campus Amsterdam, Amsterdam, the Netherlands
| | - Gordon D. Lowe
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Magdalena Zoledziewska
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, United Kingdom
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Jie Huang
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Elizabeth G. Holliday
- Public Health Stream, Hunter Medical Research Institute, and School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Mark A. McEvoy
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
| | - Dena G. Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, United States of America
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, United States of America
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - P. Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart Centre, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel, Hamburg, Germany
| | - Bruce M. Psaty
- Department of Medicine, Epidemiology, and Health Services, University of Washington, Seattle WA, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle WA, United States of America
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Eco J. C. de Geus
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, the Netherlands
- EMGO+ institute, VU University & VU medical center, Amsterdam, the Netherlands
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Harald Binder
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MS, United States of America
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Jerome I. Rotter
- Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Institute for Translational Genomics and Population Sciences, Torrance, CA, United States of America
- Division of Genomic Outcomes, Departments of Pediatrics & Medicine, Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Philipp S. Wild
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site RhineMain, Mainz, Germany
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
| | - Dorret I. Boomsma
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, the Netherlands
| | - Hugh Watkins
- Cardiovascular Medicine Dept/Radcliffe Dept of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, United States of America
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Jan W. Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, the Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands
| | - Rodney J. Scott
- Information based Medicine Program, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Paul Mitchell
- Centre for Vision Research, Department of Ophthalmology, and Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Torben Hansen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christopher J. O'Donnell
- Framingham Heart Study, Population Sciences Branch, Division of Intramural Research National Heart Lung and Blood Institute, National Institutes of Health, Framingham, MA, United States of America
- Cardiology Division, Massachusetts General Hospital, Boston, MA, United States of America
| | - Nicholas L. Smith
- Group Health Research Institute, Group Health Cooperative, Seattle WA, United States of America
- Department of Epidemiology, University of Washington, Seattle WA, United States of America
- Seattle Epidemiologic Research and Information Center, Department of Veteran Affairs Office of Research and Development, Seattle, WA, United States of America
| | - David P. Strachan
- Population Health Research Institute, St George's, University of London, London, United Kingdom
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
989
|
Haller T, Leitsalu L, Fischer K, Nuotio ML, Esko T, Boomsma DI, Kyvik KO, Spector TD, Perola M, Metspalu A. MixFit: Methodology for Computing Ancestry-Related Genetic Scores at the Individual Level and Its Application to the Estonian and Finnish Population Studies. PLoS One 2017; 12:e0170325. [PMID: 28107396 PMCID: PMC5249084 DOI: 10.1371/journal.pone.0170325] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023] Open
Abstract
Ancestry information at the individual level can be a valuable resource for personalized medicine, medical, demographical and history research, as well as for tracing back personal history. We report a new method for quantitatively determining personal genetic ancestry based on genome-wide data. Numerical ancestry component scores are assigned to individuals based on comparisons with reference populations. These comparisons are conducted with an existing analytical pipeline making use of genotype phasing, similarity matrix computation and our addition-multidimensional best fitting by MixFit. The method is demonstrated by studying Estonian and Finnish populations in geographical context. We show the main differences in the genetic composition of these otherwise close European populations and how they have influenced each other. The components of our analytical pipeline are freely available computer programs and scripts one of which was developed in house (available at: www.geenivaramu.ee/en/tools/mixfit).
Collapse
Affiliation(s)
- Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Liis Leitsalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Marja-Liisa Nuotio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Dorothea Irene Boomsma
- Vrije University, Department of Biological Psychology, Netherlands Twin Register, Amsterdam, The Netherlands
| | - Kirsten Ohm Kyvik
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Tim D. Spector
- The Department of Twin Research & Genetic Epidemiology, TwinsUK Registry, Kings College London, London, United Kingdom
| | - Markus Perola
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helskinki, Finland
| | | |
Collapse
|
990
|
de Vlaming R, Okbay A, Rietveld CA, Johannesson M, Magnusson PKE, Uitterlinden AG, van Rooij FJA, Hofman A, Groenen PJF, Thurik AR, Koellinger PD. Meta-GWAS Accuracy and Power (MetaGAP) Calculator Shows that Hiding Heritability Is Partially Due to Imperfect Genetic Correlations across Studies. PLoS Genet 2017; 13:e1006495. [PMID: 28095416 PMCID: PMC5240919 DOI: 10.1371/journal.pgen.1006495] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/17/2016] [Indexed: 11/18/2022] Open
Abstract
Large-scale genome-wide association results are typically obtained from a fixed-effects meta-analysis of GWAS summary statistics from multiple studies spanning different regions and/or time periods. This approach averages the estimated effects of genetic variants across studies. In case genetic effects are heterogeneous across studies, the statistical power of a GWAS and the predictive accuracy of polygenic scores are attenuated, contributing to the so-called ‘missing heritability’. Here, we describe the online Meta-GWAS Accuracy and Power (MetaGAP) calculator (available at www.devlaming.eu) which quantifies this attenuation based on a novel multi-study framework. By means of simulation studies, we show that under a wide range of genetic architectures, the statistical power and predictive accuracy provided by this calculator are accurate. We compare the predictions from the MetaGAP calculator with actual results obtained in the GWAS literature. Specifically, we use genomic-relatedness-matrix restricted maximum likelihood to estimate the SNP heritability and cross-study genetic correlation of height, BMI, years of education, and self-rated health in three large samples. These estimates are used as input parameters for the MetaGAP calculator. Results from the calculator suggest that cross-study heterogeneity has led to attenuation of statistical power and predictive accuracy in recent large-scale GWAS efforts on these traits (e.g., for years of education, we estimate a relative loss of 51–62% in the number of genome-wide significant loci and a relative loss in polygenic score R2 of 36–38%). Hence, cross-study heterogeneity contributes to the missing heritability. Large-scale genome-wide association studies are uncovering the genetic architecture of traits which are affected by many genetic variants. In such efforts, one typically meta-analyzes association results from multiple studies spanning different regions and/or time periods. Results from such efforts do not yet capture a large share of the heritability. The origins of this so-called ‘missing heritability’ have been strongly debated. One factor exacerbating the missing heritability is heterogeneity in the effects of genetic variants across studies. The effect of this type of heterogeneity on statistical power to detect associated genetic variants and the accuracy of polygenic predictions is poorly understood. In the current study, we derive the precise effects of heterogeneity in genetic effects across studies on both the statistical power to detect associated genetic variants as well as the accuracy of polygenic predictions. We present an online calculator, available at www.devlaming.eu, which accounts for these effects. By means of this calculator, we show that imperfect genetic correlations between studies substantially decrease statistical power and predictive accuracy and, thereby, contribute to the missing heritability. The MetaGAP calculator helps researchers to gauge how sensitive their results will be to heterogeneity in genetic effects across studies. If strong heterogeneity is expected, random-effects meta-analysis methods should be used instead of fixed-effects methods.
Collapse
Affiliation(s)
- Ronald de Vlaming
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Applied Economics, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam, the Netherlands
| | - Aysu Okbay
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam, the Netherlands
| | - Cornelius A. Rietveld
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Applied Economics, Erasmus School of Economics, Rotterdam, the Netherlands
| | - Magnus Johannesson
- Department of Economics, Stockholm School of Economics, Stockholm, Sweden
| | - Patrik K. E. Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - André G. Uitterlinden
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Frank J. A. van Rooij
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Albert Hofman
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Patrick J. F. Groenen
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Econometric Institute, Erasmus School of Economics, Rotterdam, the Netherlands
| | - A. Roy Thurik
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Applied Economics, Erasmus School of Economics, Rotterdam, the Netherlands
- Montpellier Business School, Montpellier, France
| | - Philipp D. Koellinger
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Applied Economics, Erasmus School of Economics, Rotterdam, the Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam, the Netherlands
- * E-mail:
| |
Collapse
|
991
|
Thompson PM, Andreassen OA, Arias-Vasquez A, Bearden CE, Boedhoe PS, Brouwer RM, Buckner RL, Buitelaar JK, Bulayeva KB, Cannon DM, Cohen RA, Conrod PJ, Dale AM, Deary IJ, Dennis EL, de Reus MA, Desrivieres S, Dima D, Donohoe G, Fisher SE, Fouche JP, Francks C, Frangou S, Franke B, Ganjgahi H, Garavan H, Glahn DC, Grabe HJ, Guadalupe T, Gutman BA, Hashimoto R, Hibar DP, Holland D, Hoogman M, Hulshoff Pol HE, Hosten N, Jahanshad N, Kelly S, Kochunov P, Kremen WS, Lee PH, Mackey S, Martin NG, Mazoyer B, McDonald C, Medland SE, Morey RA, Nichols TE, Paus T, Pausova Z, Schmaal L, Schumann G, Shen L, Sisodiya SM, Smit DJA, Smoller JW, Stein DJ, Stein JL, Toro R, Turner JA, van den Heuvel MP, van den Heuvel OL, van Erp TGM, van Rooij D, Veltman DJ, Walter H, Wang Y, Wardlaw JM, Whelan CD, Wright MJ, Ye J. ENIGMA and the individual: Predicting factors that affect the brain in 35 countries worldwide. Neuroimage 2017; 145:389-408. [PMID: 26658930 PMCID: PMC4893347 DOI: 10.1016/j.neuroimage.2015.11.057] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/16/2015] [Accepted: 11/23/2015] [Indexed: 11/22/2022] Open
Abstract
In this review, we discuss recent work by the ENIGMA Consortium (http://enigma.ini.usc.edu) - a global alliance of over 500 scientists spread across 200 institutions in 35 countries collectively analyzing brain imaging, clinical, and genetic data. Initially formed to detect genetic influences on brain measures, ENIGMA has grown to over 30 working groups studying 12 major brain diseases by pooling and comparing brain data. In some of the largest neuroimaging studies to date - of schizophrenia and major depression - ENIGMA has found replicable disease effects on the brain that are consistent worldwide, as well as factors that modulate disease effects. In partnership with other consortia including ADNI, CHARGE, IMAGEN and others1, ENIGMA's genomic screens - now numbering over 30,000 MRI scans - have revealed at least 8 genetic loci that affect brain volumes. Downstream of gene findings, ENIGMA has revealed how these individual variants - and genetic variants in general - may affect both the brain and risk for a range of diseases. The ENIGMA consortium is discovering factors that consistently affect brain structure and function that will serve as future predictors linking individual brain scans and genomic data. It is generating vast pools of normative data on brain measures - from tens of thousands of people - that may help detect deviations from normal development or aging in specific groups of subjects. We discuss challenges and opportunities in applying these predictors to individual subjects and new cohorts, as well as lessons we have learned in ENIGMA's efforts so far.
Collapse
Affiliation(s)
- Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA; Departments of Neurosciences, Radiology, Psychiatry, and Cognitive Science, University of California, San Diego 92093, CA, USA
| | - Ole A Andreassen
- NORMENT-KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo 0315, Norway; NORMENT-KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo 0315, Norway
| | - Alejandro Arias-Vasquez
- Donders Center for Cognitive Neuroscience, Departments of Psychiatry, Human Genetics & Cognitive Neuroscience, Radboud University Medical Center, Nijmegen 6525, The Netherlands
| | - Carrie E Bearden
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA 90095, USA; Dept. of Psychology, University of California, Los Angeles, CA 90095, USA; Brain Research Institute, University of California, Los Angeles, CA 90095, USA
| | - Premika S Boedhoe
- Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands; Department of Psychiatry, VU University Medical Center (VUMC), Amsterdam, The Netherlands; Neuroscience Campus Amsterdam, VU/VUMC, Amsterdam, The Netherlands
| | - Rachel M Brouwer
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht 3584 CX, The Netherlands
| | - Randy L Buckner
- Department of Psychiatry, Massachusetts General Hospital, Boston 02114, USA
| | - Jan K Buitelaar
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands; Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Kazima B Bulayeva
- N.I. Vavilov Institute of General Genetics, Russian Academy of Sciences, Gubkin str. 3, Moscow 119991, Russia
| | - Dara M Cannon
- National Institute of Mental Health Intramural Research Program, Bethesda 20892, USA; Neuroimaging & Cognitive Genomics Centre (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Ronald A Cohen
- Institute on Aging, University of Florida, Gainesville, FL 32611, USA
| | - Patricia J Conrod
- Department of Psychological Medicine and Psychiatry, Section of Addiction, King's College London, University of London, UK
| | - Anders M Dale
- Departments of Neurosciences, Radiology, Psychiatry, and Cognitive Science, University of California, San Diego, La Jolla, CA 92093-0841, USA
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Emily L Dennis
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA
| | - Marcel A de Reus
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht 3584 CX, The Netherlands
| | - Sylvane Desrivieres
- MRC-SGDP Centre, Institute of Psychiatry, King's College London, London SE5 8AF, UK
| | - Danai Dima
- Institute of Psychiatry, Psychology and Neuroscience, King׳s College London, UK; Clinical Neuroscience Studies (CNS) Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA
| | - Gary Donohoe
- Neuroimaging and Cognitive Genomics center (NICOG), School of Psychology, National University of Ireland, Galway, Ireland
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Jean-Paul Fouche
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Clyde Francks
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Sophia Frangou
- Clinical Neuroscience Studies (CNS) Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525, The Netherlands; Department of Psychiatry, Radboud University Medical Center, Nijmegen 6525, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Habib Ganjgahi
- Department of Statistics, The University of Warwick, Coventry, UK
| | - Hugh Garavan
- Psychiatry Department, University of Vermont, VT, USA
| | - David C Glahn
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA; Olin Neuropsychiatric Research Center, Hartford, CT 06114, USA
| | - Hans J Grabe
- Department of Psychiatry, University Medicine Greifswald, Greifswald 17489, Germany; Department of Psychiatry and Psychotherapy, HELIOS Hospital, Stralsund 18435, Germany
| | - Tulio Guadalupe
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen 6525 XD, The Netherlands; International Max Planck Research School for Language Sciences, Nijmegen 6525 XD, The Netherlands
| | - Boris A Gutman
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Japan
| | - Derrek P Hibar
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA
| | - Dominic Holland
- Departments of Neurosciences, Radiology, Psychiatry, and Cognitive Science, University of California, San Diego, La Jolla, CA 92093-0841, USA
| | - Martine Hoogman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Hilleke E Hulshoff Pol
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht 3584 CX, The Netherlands
| | - Norbert Hosten
- Department of Radiology University Medicine Greifswald, Greifswald 17475, Germany
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA
| | - Sinead Kelly
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA
| | - Peter Kochunov
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - William S Kremen
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Phil H Lee
- Center for Human Genetic Research, Massachusetts General Hospital, USA; Department of Psychiatry, Harvard Medical School, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, USA
| | - Scott Mackey
- Department of Psychiatry, University of Vermont, Burlington 05401, VT, USA
| | | | - Bernard Mazoyer
- Groupe d'imagerie Neurofonctionnelle, UMR5296 CNRS CEA Université de Bordeaux, France
| | - Colm McDonald
- Neuroimaging & Cognitive Genomics Centre (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Rajendra A Morey
- Duke Institute for Brain Sciences, Duke University, NC 27710, USA
| | - Thomas E Nichols
- Department of Statistics & WMG, University of Warwick, Coventry CV4 7AL, UK; FMRIB Centre, University of Oxford, Oxford OX3 9DU, UK
| | - Tomas Paus
- Rotman Research Institute, Baycrest, Toronto, ON, Canada; Departments of Psychology and Psychiatry, University of Toronto, Toronto, Canada; Child Mind Institute, NY, USA
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Lianne Schmaal
- Department of Psychiatry, VU University Medical Center (VUMC), Amsterdam, The Netherlands; Neuroscience Campus Amsterdam, VU/VUMC, Amsterdam, The Netherlands
| | - Gunter Schumann
- MRC-SGDP Centre, Institute of Psychiatry, King's College London, London SE5 8AF, UK
| | - Li Shen
- Center for Neuroimaging, Dept. of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th Street, Suite 4100, Indianapolis, IN 46202, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 355 W. 16th Street, Suite 4100, Indianapolis, IN 46202, USA
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London WC1N 3BG, UK and Epilepsy Society, Bucks, UK
| | - Dirk J A Smit
- Department of Biological Psychology, VU University Amsterdam, Amsterdam, The Netherlands; Neuroscience Campus Amsterdam, VU/VUMC, Amsterdam, The Netherlands
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, USA
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa; MRC Research Unit on Anxiety & Stress Disorders, South Africa
| | - Jason L Stein
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA; Neurogenetics Program, Department of Neurology, UCLA School of Medicine, Los Angeles 90095, USA
| | | | - Jessica A Turner
- Departments of Psychology and Neuroscience, Georgia State University, Atlanta, GA 30302, USA
| | - Martijn P van den Heuvel
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht 3584 CX, The Netherlands
| | - Odile L van den Heuvel
- Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands; Department of Psychiatry, VU University Medical Center (VUMC), Amsterdam, The Netherlands; Neuroscience Campus Amsterdam, VU/VUMC, Amsterdam, The Netherlands
| | - Theo G M van Erp
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92617, USA
| | - Daan van Rooij
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Dick J Veltman
- Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands; Department of Psychiatry, VU University Medical Center (VUMC), Amsterdam, The Netherlands; Neuroscience Campus Amsterdam, VU/VUMC, Amsterdam, The Netherlands
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, CCM, Berlin 10117, Germany
| | - Yalin Wang
- School of Computing, Informatics and Decision Systems Engineering, Arizona State University, AZ 85281, USA
| | - Joanna M Wardlaw
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Christopher D Whelan
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine of the University of Southern California, Marina del Rey 90292, USA
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane 4072, Australia
| | - Jieping Ye
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
992
|
|
993
|
The transcriptional architecture of phenotypic dimorphism. Nat Ecol Evol 2017; 1:6. [PMID: 28812569 DOI: 10.1038/s41559-016-0006] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/06/2016] [Indexed: 12/11/2022]
Abstract
The profound differences in gene expression between the sexes are increasingly used to study the molecular basis of sexual dimorphism, sexual selection and sexual conflict. Studies of transcriptional architecture, based on comparisons of gene expression, have also been implemented for a wide variety of other intra-specific polymorphisms. These efforts are based on key assumptions regarding the relationship between transcriptional architecture, phenotypic variation and the target of selection. Some of these assumptions are better supported by available evidence than others. In all cases, the evidence is largely circumstantial, leaving considerable gaps in our understanding of the relationship between transcriptional and phenotypic dimorphism.
Collapse
|
994
|
Kofler T, Thériault S, Bossard M, Aeschbacher S, Bernet S, Krisai P, Blum S, Risch M, Risch L, Albert CM, Paré G, Conen D. Relationships of Measured and Genetically Determined Height With the Cardiac Conduction System in Healthy Adults. Circ Arrhythm Electrophysiol 2017; 10:CIRCEP.116.004735. [DOI: 10.1161/circep.116.004735] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 11/21/2016] [Indexed: 11/16/2022]
Abstract
Background—
Increasing height is an independent risk factor for atrial fibrillation, but the underlying mechanisms are unknown. We hypothesized that height-related differences in electric conduction could be potential mediators of this relationship.
Methods and Results—
We enrolled 2149 adults aged 25 to 41 years from the general population. Height was directly measured, and a resting 12-lead ECG obtained under standardized conditions. Multivariable linear regression models were used to evaluate the association between measured height and ECG parameters. Mendelian randomization analyses were then performed using 655 independent height-associated genetic variants previously identified in the GIANT consortium. Median age was 37 years, and median height was 1.71 m. Median PR interval, QRS duration, and QTc interval were 156, 88, and 402 ms, respectively. After multivariable adjustment, β-coefficients (95% confidence intervals) per 10 cm increase in measured height were 4.17 (2.65–5.69;
P
<0.0001) for PR interval and 2.06 (1.54–2.58;
P
<0.0001) for QRS duration. Height was not associated with QTc interval or the Sokolow–Lyon index. An increase of 10 cm in genetically determined height was associated with increases of 4.33 ms (0.76–7.96;
P
=0.02) in PR interval and 2.57 ms (1.33–3.83;
P
<0.0001) in QRS duration but was not related to QTc interval or Sokolow–Lyon index.
Conclusions—
In this large population-based study, we found significant associations of measured and genetically determined height with PR interval and QRS duration. Our findings suggest that adult height is a marker of altered cardiac conduction and that these relationships may be causal.
Collapse
Affiliation(s)
- Thomas Kofler
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Sébastien Thériault
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Matthias Bossard
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Stefanie Aeschbacher
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Selina Bernet
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Philipp Krisai
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Steffen Blum
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Martin Risch
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Lorenz Risch
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Christine M. Albert
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - Guillaume Paré
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| | - David Conen
- From the Division of Internal Medicine, Department of Medicine (T.K., S.A., P.K., S.B., D.C.), Cardiovascular Research Institute Basel (T.K., M.B., S.A., S.B., P.K., S.B., D.C.), and Cardiology Division, Department of Medicine (M.B.), University Hospital Basel, Switzerland; Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute (S.T., M.B., G.P., D.C.) and Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine (S.T., G.P.),
| |
Collapse
|
995
|
Levine ME, Langfelder P, Horvath S. A Weighted SNP Correlation Network Method for Estimating Polygenic Risk Scores. Methods Mol Biol 2017; 1613:277-290. [PMID: 28849564 PMCID: PMC5998804 DOI: 10.1007/978-1-4939-7027-8_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Polygenic scores are useful for examining the joint associations of genetic markers. However, because traditional methods involve summing weighted allele counts, they may fail to capture the complex nature of biology. Here we describe a network-based method, which we call weighted SNP correlation network analysis (WSCNA), and demonstrate how it could be used to generate meaningful polygenic scores. Using data on human height in a US population of non-Hispanic whites, we illustrate how this method can be used to identify SNP networks from GWAS data, create network-specific polygenic scores, examine network topology to identify hub SNPs, and gain biological insights into complex traits. In our example, we show that this method explains a larger proportion of the variance in human height than traditional polygenic score methods. We also identify hub genes and pathways that have previously been identified as influencing human height. In moving forward, this method may be useful for generating genetic susceptibility measures for other health related traits, examining genetic pleiotropy, identifying at-risk individuals, examining gene score by environmental effects, and gaining a deeper understanding of the underlying biology of complex traits.
Collapse
Affiliation(s)
- Morgan E Levine
- Department of Human Genetics, University of California, Box 708822, 695 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, 90095, USA.
| | - Peter Langfelder
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, 90095, USA
| | - Steve Horvath
- Department of Human Genetics, University of California, Box 708822, 695 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
- Department of Biostatistics, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
996
|
Lotta LA, Gulati P, Day FR, Payne F, Ongen H, van de Bunt M, Gaulton KJ, Eicher JD, Sharp SJ, Luan J, De Lucia Rolfe E, Stewart ID, Wheeler E, Willems SM, Adams C, Yaghootkar H, EPIC-InterAct Consortium, Cambridge FPLD1 Consortium, Forouhi NG, Khaw KT, Johnson AD, Semple RK, Frayling T, Perry JRB, Dermitzakis E, McCarthy MI, Barroso I, Wareham NJ, Savage DB, Langenberg C, O’Rahilly S, Scott RA. Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance. Nat Genet 2017; 49:17-26. [PMID: 27841877 PMCID: PMC5774584 DOI: 10.1038/ng.3714] [Citation(s) in RCA: 422] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023]
Abstract
Insulin resistance is a key mediator of obesity-related cardiometabolic disease, yet the mechanisms underlying this link remain obscure. Using an integrative genomic approach, we identify 53 genomic regions associated with insulin resistance phenotypes (higher fasting insulin levels adjusted for BMI, lower HDL cholesterol levels and higher triglyceride levels) and provide evidence that their link with higher cardiometabolic risk is underpinned by an association with lower adipose mass in peripheral compartments. Using these 53 loci, we show a polygenic contribution to familial partial lipodystrophy type 1, a severe form of insulin resistance, and highlight shared molecular mechanisms in common/mild and rare/severe insulin resistance. Population-level genetic analyses combined with experiments in cellular models implicate CCDC92, DNAH10 and L3MBTL3 as previously unrecognized molecules influencing adipocyte differentiation. Our findings support the notion that limited storage capacity of peripheral adipose tissue is an important etiological component in insulin-resistant cardiometabolic disease and highlight genes and mechanisms underpinning this link.
Collapse
Affiliation(s)
- Luca A. Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Pawan Gulati
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Felix R. Day
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Felicity Payne
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United
Kingdom
| | - Halit Ongen
- Department of Genetic Medicine and Development, University of Geneva
Medical School, Geneva, Switzerland
| | - Martijn van de Bunt
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University
of Oxford, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford,
Oxford, United Kingdom
| | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La
Jolla, USA
| | - John D. Eicher
- Population Sciences Branch, Division of Intramural Research,
National Heart, Lung and Blood Institute, Bethesda, USA
| | - Stephen J. Sharp
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Jian’an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | | | - Isobel D. Stewart
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Eleanor Wheeler
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United
Kingdom
| | - Sara M. Willems
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Claire Adams
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, Institute of Biomedical and Clinical
Science, University of Exeter Medical School, Royal Devon and Exeter Hospital,
Exeter, United Kingdom
| | | | | | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of
Cambridge, Cambridge, United Kingdom
| | - Andrew D. Johnson
- Population Sciences Branch, Division of Intramural Research,
National Heart, Lung and Blood Institute, Bethesda, USA
| | - Robert K. Semple
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Timothy Frayling
- Genetics of Complex Traits, Institute of Biomedical and Clinical
Science, University of Exeter Medical School, Royal Devon and Exeter Hospital,
Exeter, United Kingdom
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Emmanouil Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva
Medical School, Geneva, Switzerland
| | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University
of Oxford, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford,
Oxford, United Kingdom
| | - Inês Barroso
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United
Kingdom
| | | | - David B. Savage
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Stephen O’Rahilly
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| |
Collapse
|
997
|
Zheng J, Rodriguez S, Laurin C, Baird D, Trela-Larsen L, Erzurumluoglu MA, Zheng Y, White J, Giambartolomei C, Zabaneh D, Morris R, Kumari M, Casas JP, Hingorani AD, UCLEB Consortium, Evans DM, Gaunt TR, Day INM. HAPRAP: a haplotype-based iterative method for statistical fine mapping using GWAS summary statistics. Bioinformatics 2017; 33:79-86. [PMID: 27591082 PMCID: PMC5544112 DOI: 10.1093/bioinformatics/btw565] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/29/2016] [Accepted: 08/26/2016] [Indexed: 11/21/2022] Open
Abstract
MOTIVATION Fine mapping is a widely used approach for identifying the causal variant(s) at disease-associated loci. Standard methods (e.g. multiple regression) require individual level genotypes. Recent fine mapping methods using summary-level data require the pairwise correlation coefficients ([Formula: see text]) of the variants. However, haplotypes rather than pairwise [Formula: see text], are the true biological representation of linkage disequilibrium (LD) among multiple loci. In this article, we present an empirical iterative method, HAPlotype Regional Association analysis Program (HAPRAP), that enables fine mapping using summary statistics and haplotype information from an individual-level reference panel. RESULTS Simulations with individual-level genotypes show that the results of HAPRAP and multiple regression are highly consistent. In simulation with summary-level data, we demonstrate that HAPRAP is less sensitive to poor LD estimates. In a parametric simulation using Genetic Investigation of ANthropometric Traits height data, HAPRAP performs well with a small training sample size (N < 2000) while other methods become suboptimal. Moreover, HAPRAP's performance is not affected substantially by single nucleotide polymorphisms (SNPs) with low minor allele frequencies. We applied the method to existing quantitative trait and binary outcome meta-analyses (human height, QTc interval and gallbladder disease); all previous reported association signals were replicated and two additional variants were independently associated with human height. Due to the growing availability of summary level data, the value of HAPRAP is likely to increase markedly for future analyses (e.g. functional prediction and identification of instruments for Mendelian randomization). AVAILABILITY AND IMPLEMENTATION The HAPRAP package and documentation are available at http://apps.biocompute.org.uk/haprap/ CONTACT: : jie.zheng@bristol.ac.uk or tom.gaunt@bristol.ac.ukSupplementary information: Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jie Zheng
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Santiago Rodriguez
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Charles Laurin
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Denis Baird
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, Bristol, UK
| | - Lea Trela-Larsen
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Mesut A Erzurumluoglu
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- Department of Health Sciences, Genetic Epidemiology Group, University of Leicester, Leicester, UK
| | - Yi Zheng
- Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, USA
| | - Jon White
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London, UK
| | - Claudia Giambartolomei
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London, UK
| | - Delilah Zabaneh
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London, UK
| | - Richard Morris
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Meena Kumari
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London, UK
| | - Juan P Casas
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London, UK
- Department of Primary Care & Population Health, University College London, Royal Free Campus, London, UK
| | - Aroon D Hingorani
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London, UK
- Centre for Clinical Pharmacology, University College London, London, UK, Division of Medicine
| | | | - David M Evans
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, Bristol, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia, QLD
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Ian N M Day
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
998
|
Iacono WG, Malone SM, Vrieze SI. Endophenotype best practices. Int J Psychophysiol 2017; 111:115-144. [PMID: 27473600 PMCID: PMC5219856 DOI: 10.1016/j.ijpsycho.2016.07.516] [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: 04/11/2016] [Revised: 07/21/2016] [Accepted: 07/24/2016] [Indexed: 01/19/2023]
Abstract
This review examines the current state of electrophysiological endophenotype research and recommends best practices that are based on knowledge gleaned from the last decade of molecular genetic research with complex traits. Endophenotype research is being oversold for its potential to help discover psychopathology relevant genes using the types of small samples feasible for electrophysiological research. This is largely because the genetic architecture of endophenotypes appears to be very much like that of behavioral traits and disorders: they are complex, influenced by many variants (e.g., tens of thousands) within many genes, each contributing a very small effect. Out of over 40 electrophysiological endophenotypes covered by our review, only resting heart, a measure that has received scant advocacy as an endophenotype, emerges as an electrophysiological variable with verified associations with molecular genetic variants. To move the field forward, investigations designed to discover novel variants associated with endophenotypes will need extremely large samples best obtained by forming consortia and sharing data obtained from genome wide arrays. In addition, endophenotype research can benefit from successful molecular genetic studies of psychopathology by examining the degree to which these verified psychopathology-relevant variants are also associated with an endophenotype, and by using knowledge about the functional significance of these variants to generate new endophenotypes. Even without molecular genetic associations, endophenotypes still have value in studying the development of disorders in unaffected individuals at high genetic risk, constructing animal models, and gaining insight into neural mechanisms that are relevant to clinical disorder.
Collapse
|
999
|
Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D, Manolagas SC. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol Rev 2017. [PMID: 27807202 DOI: 10.1152/physrev.00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Michaël R Laurent
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Vanessa Dubois
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Frank Claessens
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Charles A O'Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Roger Bouillon
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Dirk Vanderschueren
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| |
Collapse
|
1000
|
Franck SE, Broer L, van der Lely AJ, Kamenicky P, Bernabéu I, Malchiodi E, Delhanty PJ, Rivadeneira F, Neggers SJ. The Effect of the Exon-3-Deleted Growth Hormone Receptor on Pegvisomant-Treated Acromegaly: A Systematic Review and Meta-Analysis. Neuroendocrinology 2017; 105:131-140. [PMID: 27513761 PMCID: PMC5637298 DOI: 10.1159/000448844] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/02/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND The common exon 3 deletion polymorphism of the growth hormone receptor (d3-GHR) is associated with disease severity in acromegaly patients. The GHR antagonist pegvisomant (PEGV) is highly effective in treating severe acromegaly. Response to PEGV treatment seems to be influenced by d3-GHR and appears to be more responsive to PEGV, although available results remain conflicting. OBJECTIVE To assess the influence of d3-GHR on the responsiveness of acromegaly patients to PEGV by compiling the evidence derived from the largest available studies. DESIGN A systematic review of the literature identified three published studies and one conference abstract. Acromegaly patients (n = 324, 49.7% d3-GHR carriers) were treated with either PEGV monotherapy or PEGV combined with long-acting somatostatin analogues and/or cabergoline. A meta-analysis of raw data from these studies was performed. RESULTS No significant effect of the d3-GHR was observed while bringing insulin-like growth factor I (IGF-I) levels below the upper limit of normal with PEGV, which was defined as the lowest IGF-I level during PEGV treatment (mean difference: -2.3%; 95% CI: -6.5 to 1.8, p = 0.270). The PEGV dose required to achieve the lowest IGF-I levels was also not significantly influenced by individuals carrying d3-GHR (mean difference: 4.1 mg weekly; 95% CI: -5.1 to 13.2, p = 0.385). For both outcomes, separate analysis of PEGV monotherapy and combination treatment gave similar results. CONCLUSION Our findings suggest that the d3-GHR polymorphism has no effect on biochemical disease control in acromegaly, as it is not of added value for either the prediction of PEGV responsiveness or the determination of the required PEGV dose.
Collapse
Affiliation(s)
- Sanne E. Franck
- Department of Internal Medicine, Endocrinology Section Rotterdam, Rotterdam, The Netherlands
- *S.E. Franck, Department of Internal Medicine, Endocrinology Section, Erasmus University Medical Center, PO Box 2040, NL-3000 CA Rotterdam (The Netherlands), E-Mail
| | - Linda Broer
- Department of Internal Medicine, Endocrinology Section Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aart Jan van der Lely
- Department of Internal Medicine, Endocrinology Section Rotterdam, Rotterdam, The Netherlands
| | - Peter Kamenicky
- Department of Assistance Publique-Hôpitaux de Paris, Service d'Endocrinologie et des Maladies de la Reproduction, Hôpital de Bicêtre, Paris, France
- Department of Faculté de Médecine, UMR-S1185, Université Paris-Sud, Le Kremlin Bicêtre, Paris, France
- Department of Institut National de la Santé et de la Recherche Médicale (INSERM) U1185, Paris, France
| | - Ignacio Bernabéu
- Department of Endocrinology, Clinic University Hospital - Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Elena Malchiodi
- Department of Department of Clinical Sciences and Community Health, Endocrinology and Diabetology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Patric J.D. Delhanty
- Department of Internal Medicine, Endocrinology Section Rotterdam, Rotterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Endocrinology Section Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | |
Collapse
|