scispace - formally typeset
Search or ask a question

Showing papers by "Michael Boehnke published in 2019"


Journal ArticleDOI
Eli A. Stahl1, Eli A. Stahl2, Gerome Breen3, Andreas J. Forstner  +339 moreInstitutions (107)
TL;DR: Genome-wide analysis identifies 30 loci associated with bipolar disorder, allowing for comparisons of shared genes and pathways with other psychiatric disorders, including schizophrenia and depression.
Abstract: Bipolar disorder is a highly heritable psychiatric disorder. We performed a genome-wide association study (GWAS) including 20,352 cases and 31,358 controls of European descent, with follow-up analysis of 822 variants with P < 1 × 10-4 in an additional 9,412 cases and 137,760 controls. Eight of the 19 variants that were genome-wide significant (P < 5 × 10-8) in the discovery GWAS were not genome-wide significant in the combined analysis, consistent with small effect sizes and limited power but also with genetic heterogeneity. In the combined analysis, 30 loci were genome-wide significant, including 20 newly identified loci. The significant loci contain genes encoding ion channels, neurotransmitter transporters and synaptic components. Pathway analysis revealed nine significantly enriched gene sets, including regulation of insulin secretion and endocannabinoid signaling. Bipolar I disorder is strongly genetically correlated with schizophrenia, driven by psychosis, whereas bipolar II disorder is more strongly correlated with major depressive disorder. These findings address key clinical questions and provide potential biological mechanisms for bipolar disorder.

1,090 citations


Posted ContentDOI
Daniel Taliun1, Daniel N. Harris2, Michael D. Kessler2, Jedidiah Carlson3  +191 moreInstitutions (61)
06 Mar 2019-bioRxiv
TL;DR: The nearly complete catalog of genetic variation in TOPMed studies provides unique opportunities for exploring the contributions of rare and non-coding sequence variants to phenotypic variation as well as resources and early insights from the sequence data.
Abstract: Summary paragraph The Trans-Omics for Precision Medicine (TOPMed) program seeks to elucidate the genetic architecture and disease biology of heart, lung, blood, and sleep disorders, with the ultimate goal of improving diagnosis, treatment, and prevention. The initial phases of the program focus on whole genome sequencing of individuals with rich phenotypic data and diverse backgrounds. Here, we describe TOPMed goals and design as well as resources and early insights from the sequence data. The resources include a variant browser, a genotype imputation panel, and sharing of genomic and phenotypic data via dbGaP. In 53,581 TOPMed samples, >400 million single-nucleotide and insertion/deletion variants were detected by alignment with the reference genome. Additional novel variants are detectable through assembly of unmapped reads and customized analysis in highly variable loci. Among the >400 million variants detected, 97% have frequency

662 citations


Journal ArticleDOI
Ayush Giri1, Jacklyn N. Hellwege2, Jacob M. Keaton2, Jacob M. Keaton1, Jihwan Park3, Chengxiang Qiu3, Helen R. Warren4, Helen R. Warren5, Eric S. Torstenson1, Eric S. Torstenson2, Csaba P. Kovesdy6, Yan V. Sun7, Otis D. Wilson2, Otis D. Wilson1, Cassianne Robinson-Cohen1, Christianne L. Roumie1, Cecilia P. Chung1, K A Birdwell1, K A Birdwell6, Scott M. Damrauer6, Scott L. DuVall, Derek Klarin, Kelly Cho8, Yu Wang1, Evangelos Evangelou9, Evangelos Evangelou10, Claudia P. Cabrera4, Claudia P. Cabrera5, Louise V. Wain5, Louise V. Wain11, Rojesh Shrestha3, Brian S. Mautz1, Elvis A. Akwo1, Muralidharan Sargurupremraj12, Stéphanie Debette12, Michael Boehnke13, Laura J. Scott13, Jian'an Luan14, Zhao J-H.14, Sara M. Willems14, Sébastien Thériault15, Nabi Shah16, Nabi Shah17, Christopher Oldmeadow18, Peter Almgren19, Ruifang Li-Gao20, Niek Verweij21, Thibaud Boutin22, Massimo Mangino23, Massimo Mangino24, Ioanna Ntalla4, Elena V. Feofanova25, Praveen Surendran14, James P. Cook26, Savita Karthikeyan14, Najim Lahrouchi27, Ching-Ti Liu28, Nuno Sepúlveda29, Tom G. Richardson30, Aldi T. Kraja31, Philippe Amouyel32, Martin Farrall33, Neil Poulter9, Markku Laakso34, Eleftheria Zeggini35, Peter S. Sever36, Robert A. Scott14, Claudia Langenberg14, Nicholas J. Wareham14, David Conen37, Palmer Cna.17, John Attia18, Daniel I. Chasman38, Paul M. Ridker38, Olle Melander19, Dennis O. Mook-Kanamori20, Harst Pvd.21, Francesco Cucca39, David Schlessinger36, Caroline Hayward22, Tim D. Spector23, Jarvelin M-R.1, Branwen J. Hennig40, Branwen J. Hennig29, Nicholas J. Timpson30, Wei W-Q.1, J C Smith1, Yaomin Xu1, Michael E. Matheny, E E Siew1, C M Lindgren33, C M Lindgren27, C M Lindgren41, Herzig K-H., George Dedoussis42, Josh C. Denny1, Bruce M. Psaty43, Howson Jmm.14, Patricia B. Munroe5, Patricia B. Munroe4, Christopher Newton-Cheh44, Mark J. Caulfield5, Mark J. Caulfield4, Paul Elliott5, Paul Elliott9, J M Gaziano45, J M Gaziano46, John Concato, Wilson Pwf.6, Philip S. Tsao45, D.R. Velez Edwards1, D.R. Velez Edwards2, Katalin Susztak3, Christopher J. O'Donnell38, Adriana M. Hung2, Adriana M. Hung1, Todd L. Edwards1, Todd L. Edwards2 
TL;DR: Analysis of blood pressure data from the Million Veteran Program trans-ethnic cohort identifies common and rare variants, and genetically predicted gene expression across multiple tissues associated with systolic, diastolic and pulse pressure in over 775,000 individuals.
Abstract: In this trans-ethnic multi-omic study, we reinterpret the genetic architecture of blood pressure to identify genes, tissues, phenomes and medication contexts of blood pressure homeostasis. We discovered 208 novel common blood pressure SNPs and 53 rare variants in genome-wide association studies of systolic, diastolic and pulse pressure in up to 776,078 participants from the Million Veteran Program (MVP) and collaborating studies, with analysis of the blood pressure clinical phenome in MVP. Our transcriptome-wide association study detected 4,043 blood pressure associations with genetically predicted gene expression of 840 genes in 45 tissues, and mouse renal single-cell RNA sequencing identified upregulated blood pressure genes in kidney tubule cells.

310 citations


01 Jan 2019
TL;DR: Trans-ancestry meta-analysis of estimated glomerular filtration rate (eGFR) from 1,046,070 individuals identifies 264 associated loci, providing a resource of molecular targets for translational research of chronic kidney disease.
Abstract: Chronic kidney disease (CKD) is responsible for a public health burden with multi-systemic complications. Through trans-ancestry meta-analysis of genome-wide association studies of estimated glomerular filtration rate (eGFR) and independent replication (n = 1,046,070), we identified 264 associated loci (166 new). Of these, 147 were likely to be relevant for kidney function on the basis of associations with the alternative kidney function marker blood urea nitrogen (n = 416,178). Pathway and enrichment analyses, including mouse models with renal phenotypes, support the kidney as the main target organ. A genetic risk score for lower eGFR was associated with clinically diagnosed CKD in 452,264 independent individuals. Colocalization analyses of associations with eGFR among 783,978 European-ancestry individuals and gene expression across 46 human tissues, including tubulo-interstitial and glomerular kidney compartments, identified 17 genes differentially expressed in kidney. Fine-mapping highlighted missense driver variants in 11 genes and kidney-specific regulatory variants. These results provide a comprehensive priority list of molecular targets for translational research.Trans-ancestry meta-analysis of estimated glomerular filtration rate (eGFR) from 1,046,070 individuals identifies 264 associated loci, providing a resource of molecular targets for translational research of chronic kidney disease.

243 citations


Journal ArticleDOI
Niamh Mullins1, Tim B. Bigdeli2, Anders D. Børglum3, Jonathan R. I. Coleman3, Ditte Demontis4, Divya Mehta4, Robert Power5, Stephan Ripke6, Eli A. Stahl7, Anna Starnawska8, Adebayo Anjorin9, M.R.C.Psych10, A. Corvin2, Alan R. Sanders11, Andreas J. Forstner12, Andreas Reif13, Anna C. Koller14, Beata Świątkowska15, Bernhard T. Baune16, Bertram Müller-Myhsok15, Brenda W.J.H. Penninx3, Carlos N. Pato17, Clement C. Zai18, Dan Rujescu19, David M. Hougaard20, Digby Quested21, Douglas F. Levinson22, Elisabeth B. Binder23, Enda M. Byrne, Esben Agerbo24, Fabian Streit25, Fermín Mayoral3, Frank Bellivier3, Franziska Degenhardt26, Gerome Breen27, Gunnar Morken28, Gustavo Turecki3, Guy A. Rouleau3, Hans Joergen Grabe3, Henry Völzke3, I. Jones3, Ina Giegling3, Ingrid Agartz3, Ingrid Melle3, Jacob Lawrence3, James T.R. Walters3, Jana Strohmaier3, Jianxin Shi3, Joanna Hauser3, Joanna M. Biernacka3, John B. Vincent3, John R. Kelsoe3, John Strauss3, Jolanta Lissowska3, Jonathan Pimm3, Jordan W. Smoller3, Jose Guzman-Parra3, Klaus Berger3, Laura J. Scott3, Lisa Jones3, M. Helena Azevedo3, Maciej Trzaskowski3, Manolis Kogevinas3, Marcella Rietschel3, Marco Boks3, Marcus Ising23, Maria Grigoroiu-Serbanescu3, Marian L. Hamshere3, Marion Leboyer3, Mark Frye3, Markus M. Nöthen3, Martin Alda3, Martin Preisig3, Merete Nordentoft3, Michael Boehnke3, Michael Conlon O'Donovan3, Michael John Owen3, Michele T. Pato3, Miguel E. Rentería3, Monika Budde3, Myrna M. Weissman3, Naomi R. Wray3, Nicholas Bass3, Nicholas Craddock3, Olav B. Smeland3, Ole A. Andreassen3, Ole Mors3, PV Gejman3, Pamela Sklar3, Patrick McGrath3, Per Hoffmann3, P. McGuffin3, Phil H. Lee3, Preben Bo Mortensen3, René S. Kahn3, Roel A. Ophoff3, Rolf Adolfsson3, Sandra Van der Auwera3, Srdjan Djurovic3, Stefan Kloiber3, Stefanie Heilmann-Heimbach3, Stéphane Jamain3, Steven P. Hamilton3, Susan L. McElroy3, Susanne Lucae3, Sven Cichon3, Thomas G. Schulze3, Thomas Hansen3, Thomas Werge3, Tracy M. Air3, Vishwajit Nimgaonkar3, Vivek Appadurai3, Wiepke Cahn3, Yuri Milaneschi3, Ayman H. Fanous3, Kenneth S. Kendler3, Andrew McQuillin3, Cathryn M. Lewis3 
TL;DR: This study provides new information on genetic associations and demonstrates that genetic liability for major depression increases risk for suicide attempt across psychiatric disorders.
Abstract: OBJECTIVE: More than 90% of people who attempt suicide have a psychiatric diagnosis; however, twin and family studies suggest that the genetic etiology of suicide attempt is partially distinct from that of the psychiatric disorders themselves. The authors present the largest genome-wide association study (GWAS) on suicide attempt, using cohorts of individuals with major depressive disorder, bipolar disorder, and schizophrenia from the Psychiatric Genomics Consortium. METHODS: The samples comprised 1,622 suicide attempters and 8,786 nonattempters with major depressive disorder; 3,264 attempters and 5,500 nonattempters with bipolar disorder; and 1,683 attempters and 2,946 nonattempters with schizophrenia. A GWAS on suicide attempt was performed by comparing attempters to nonattempters with each disorder, followed by a meta-analysis across disorders. Polygenic risk scoring was used to investigate the genetic relationship between suicide attempt and the psychiatric disorders. RESULTS: Three genome-wide significant loci for suicide attempt were found: one associated with suicide attempt in major depressive disorder, one associated with suicide attempt in bipolar disorder, and one in the meta-analysis of suicide attempt in mood disorders. These associations were not replicated in independent mood disorder cohorts from the UK Biobank and iPSYCH. No significant associations were found in the meta-analysis of all three disorders. Polygenic risk scores for major depression were significantly associated with suicide attempt in major depressive disorder (R2=0.25%), bipolar disorder (R2=0.24%), and schizophrenia (R2=0.40%). CONCLUSIONS: This study provides new information on genetic associations and demonstrates that genetic liability for major depression increases risk for suicide attempt across psychiatric disorders. Further collaborative efforts to increase sample size may help to robustly identify genetic associations and provide biological insights into the etiology of suicide attempt.

161 citations


Posted ContentDOI
Cassandra N. Spracklen1, Momoko Horikoshi, Young Jin Kim, Kuang Lin2, Fiona Bragg2, Sanghoon Moon, Ken Suzuki, Claudia H. T. Tam3, Yasuharu Tabara4, Soo Heon Kwak5, Fumihiko Takeuchi, Jirong Long6, Victor Jun Yu Lim7, Jin-Fang Chai7, Chien-Hsiun Chen8, Masahiro Nakatochi9, Jie Yao10, Hyeok Sun Choi11, Apoorva K Iyengar1, Hannah J Perrin1, Sarah M Brotman1, Martijn van de Bunt2, Anna L. Gloyn2, Jennifer E. Below6, Michael Boehnke12, Donald W. Bowden13, John C. Chambers14, Anubha Mahajan2, Mark I. McCarthy2, Maggie C.Y. Ng13, Lauren E. Petty6, Weihua Zhang15, Andrew P. Morris16, Linda S. Adair1, Zheng Bian17, Juliana C.N. Chan3, Li-Ching Chang8, Miao-Li Chee, Yii-Der Ida Chen10, Yuan-Tsong Chen8, Zhengming Chen2, Lee-Ming Chuang18, Shufa Du1, Penny Gordon-Larsen1, Myron D. Gross19, Xiuqing Guo10, Yu Guo17, Sohee Han, Annie-Green Howard1, Wei Huang20, Yi-Jen Hung21, Mi Yeong Hwang, Chii-Min Hwu22, Sahoko Ichihara23, Masato Isono23, Hye-Mi Jang, Guozhi Jiang3, Jost B. Jonas24, Yoichiro Kamatani25, Tomohiro Katsuya26, Takahisa Kawaguchi4, Chiea Chuen Khor27, Katsuhiko Kohara28, Myung-Shik Lee29, Nanette R. Lee30, Liming Li31, Jianjun Liu27, Andrea O.Y. Luk3, Jun Lv31, Yukinori Okada26, Mark A Pereira19, Charumathi Sabanayagam7, Shi Jinxiu20, Dong Mun Shin, Wing-Yee So3, Atsushi Takahashi, Brian Tomlinson3, Fuu Jen Tsai32, Rob M. van Dam7, Yong-Bing Xiang33, Ken Yamamoto34, Toshimasa Yamauchi25, Kyungheon Yoon, Canqing Yu31, Jian-Min Yuan35, Liang Zhang, Wei Zheng6, Michiya Igase28, Yoon Shin Cho11, Jerome I. Rotter10, Ya Xing Wang36, Wayne Huey-Herng Sheu37, Wayne Huey-Herng Sheu38, Mitsuhiro Yokota34, Jer-Yuarn Wu8, Ching-Yu Cheng7, Tien Yin Wong7, Xiao-Ou Shu6, Norihiro Kato, Kyong-Soo Park5, E-Shyong Tai7, Fumihiko Matsuda4, Woon-Puay Koh7, Ronald Cw Ma3, Shiro Maeda39, Iona Y Millwood2, Ju Young Lee, Takashi Kadowaki25, Robin G. Walters2, Bong-Jo Kim, Karen L. Mohlke1, Xueling Sim7 
28 Jun 2019-bioRxiv
TL;DR: The largest meta-analysis of East Asian individuals to identify new genetic associations and provide insight into T2D pathogenesis is performed.
Abstract: Meta-analyses of genome-wide association studies (GWAS) have identified >240 loci associated with type 2 diabetes (T2D), however most loci have been identified in analyses of European-ancestry individuals. To examine T2D risk in East Asian individuals, we meta-analyzed GWAS data in 77,418 cases and 356,122 controls. In the main analysis, we identified 298 distinct association signals at 178 loci, and across T2D association models with and without consideration of body mass index and sex, we identified 56 loci newly implicated in T2D predisposition. Common variants associated with T2D in both East Asian and European populations exhibited strongly correlated effect sizes. New associations include signals in/near GDAP1 , PTF1A , SIX3 , ALDH2 , a microRNA cluster, and genes that affect muscle and adipose differentiation. At another locus, eQTLs at two overlapping T2D signals act through two genes, NKX6-3 and ANK1 , in different tissues. Association studies in diverse populations identify additional loci and elucidate disease genes, biology, and pathways.

137 citations


01 Jan 2019
TL;DR: The authors used exome-sequencing analyses of a large cohort of patients with Type 2 diabetes and control individuals without diabetes from five ancestries to identify gene-level associations of rare variants that are associated with type 2 diabetes.
Abstract: Protein-coding genetic variants that strongly affect disease risk can yield relevant clues to disease pathogenesis. Here we report exome-sequencing analyses of 20,791 individuals with type 2 diabetes (T2D) and 24,440 non-diabetic control participants from 5 ancestries. We identify gene-level associations of rare variants (with minor allele frequencies of less than 0.5%) in 4 genes at exome-wide significance, including a series of more than 30 SLC30A8 alleles that conveys protection against T2D, and in 12 gene sets, including those corresponding to T2D drug targets (P = 6.1 × 10−3) and candidate genes from knockout mice (P = 5.2 × 10−3). Within our study, the strongest T2D gene-level signals for rare variants explain at most 25% of the heritability of the strongest common single-variant signals, and the gene-level effect sizes of the rare variants that we observed in established T2D drug targets will require 75,000–185,000 sequenced cases to achieve exome-wide significance. We propose a method to interpret these modest rare-variant associations and to incorporate these associations into future target or gene prioritization efforts.Exome-sequencing analyses of a large cohort of patients with type 2 diabetes and control individuals without diabetes from five ancestries are used to identify gene-level associations of rare variants that are associated with type 2 diabetes.

107 citations


Journal ArticleDOI
TL;DR: In this paper, Mendelian randomization (MR) and mediation techniques were used to predict 213 causal relationships between expression and DNA methylation, approximately two-thirds of which predict methylation to causally influence expression.
Abstract: We integrate comeasured gene expression and DNA methylation (DNAme) in 265 human skeletal muscle biopsies from the FUSION study with >7 million genetic variants and eight physiological traits: height, waist, weight, waist-hip ratio, body mass index, fasting serum insulin, fasting plasma glucose, and type 2 diabetes. We find hundreds of genes and DNAme sites associated with fasting insulin, waist, and body mass index, as well as thousands of DNAme sites associated with gene expression (eQTM). We find that controlling for heterogeneity in tissue/muscle fiber type reduces the number of physiological trait associations, and that long-range eQTMs (>1 Mb) are reduced when controlling for tissue/muscle fiber type or latent factors. We map genetic regulators (quantitative trait loci; QTLs) of expression (eQTLs) and DNAme (mQTLs). Using Mendelian randomization (MR) and mediation techniques, we leverage these genetic maps to predict 213 causal relationships between expression and DNAme, approximately two-thirds of which predict methylation to causally influence expression. We use MR to integrate FUSION mQTLs, FUSION eQTLs, and GTEx eQTLs for 48 tissues with genetic associations for 534 diseases and quantitative traits. We identify hundreds of genes and thousands of DNAme sites that may drive the reported disease/quantitative trait genetic associations. We identify 300 gene expression MR associations that are present in both FUSION and GTEx skeletal muscle and that show stronger evidence of MR association in skeletal muscle than other tissues, which may partially reflect differences in power across tissues. As one example, we find that increased RXRA muscle expression may decrease lean tissue mass.

94 citations


01 Jan 2019
TL;DR: In this paper, the role of rare coding variants in clinically relevant quantitative cardiometabolic traits was investigated using exome-sequencing of nearly 20,000 individuals from these regions.
Abstract: Exome-sequencing studies have generally been underpowered to identify deleterious alleles with a large effect on complex traits as such alleles are mostly rare. Because the population of northern and eastern Finland has expanded considerably and in isolation following a series of bottlenecks, individuals of these populations have numerous deleterious alleles at a relatively high frequency. Here, using exome sequencing of nearly 20,000 individuals from these regions, we investigate the role of rare coding variants in clinically relevant quantitative cardiometabolic traits. Exome-wide association studies for 64 quantitative traits identified 26 newly associated deleterious alleles. Of these 26 alleles, 19 are either unique to or more than 20 times more frequent in Finnish individuals than in other Europeans and show geographical clustering comparable to Mendelian disease mutations that are characteristic of the Finnish population. We estimate that sequencing studies of populations without this unique history would require hundreds of thousands to millions of participants to achieve comparable association power.Exome-wide sequencing studies of populations in Finland identified 26 deleterious alleles associated with 64 quantitative traits that are clinically relevant to cardiovascular and metabolic diseases.

93 citations


Journal ArticleDOI
Paul S. de Vries1, Michael R. Brown1, Amy R. Bentley2, Yun J. Sung3  +290 moreInstitutions (88)
TL;DR: In this paper, gene-alcohol interactions were incorporated into a multiancestry genome-wide association study of levels of high-density lipoprotein cholesterol, low-density cholesterol, and triglycerides.
Abstract: A person's lipid profile is influenced by genetic variants and alcohol consumption, but the contribution of interactions between these exposures has not been studied. We therefore incorporated gene-alcohol interactions into a multiancestry genome-wide association study of levels of high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and triglycerides. We included 45 studies in stage 1 (genome-wide discovery) and 66 studies in stage 2 (focused follow-up), for a total of 394,584 individuals from 5 ancestry groups. Analyses covered the period July 2014-November 2017. Genetic main effects and interaction effects were jointly assessed by means of a 2-degrees-of-freedom (df) test, and a 1-df test was used to assess the interaction effects alone. Variants at 495 loci were at least suggestively associated (P < 1 × 10-6) with lipid levels in stage 1 and were evaluated in stage 2, followed by combined analyses of stage 1 and stage 2. In the combined analysis of stages 1 and 2, a total of 147 independent loci were associated with lipid levels at P < 5 × 10-8 using 2-df tests, of which 18 were novel. No genome-wide-significant associations were found testing the interaction effect alone. The novel loci included several genes (proprotein convertase subtilisin/kexin type 5 (PCSK5), vascular endothelial growth factor B (VEGFB), and apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (APOBEC1) complementation factor (A1CF)) that have a putative role in lipid metabolism on the basis of existing evidence from cellular and experimental models.

79 citations


Journal ArticleDOI
David W. Clark1, Yukinori Okada2, Kristjan H. S. Moore3, Dan Mason  +493 moreInstitutions (142)
TL;DR: In this paper, the authors used genomic inbreeding coefficients (FROH) for >1.4 million individuals and found that FROH is significantly associated with apparently deleterious changes in 32 out of 100 traits analysed.
Abstract: In many species, the offspring of related parents suffer reduced reproductive success, a phenomenon known as inbreeding depression. In humans, the importance of this effect has remained unclear, partly because reproduction between close relatives is both rare and frequently associated with confounding social factors. Here, using genomic inbreeding coefficients (FROH) for >1.4 million individuals, we show that FROH is significantly associated (p < 0.0005) with apparently deleterious changes in 32 out of 100 traits analysed. These changes are associated with runs of homozygosity (ROH), but not with common variant homozygosity, suggesting that genetic variants associated with inbreeding depression are predominantly rare. The effect on fertility is striking: FROH equivalent to the offspring of first cousins is associated with a 55% decrease [95% CI 44-66%] in the odds of having children. Finally, the effects of FROH are confirmed within full-sibling pairs, where the variation in FROH is independent of all environmental confounding.

Journal ArticleDOI
TL;DR: In this article, the role of physical activity interactions in the genetic contribution to blood lipid levels was investigated, showing that higher levels of physical activities enhance the HDL cholesterol-increasing effects of the CLASP1, LHX1, and SNTA1 loci and attenuate the LDL cholesterol -increasing effect of the CNTNAP2 locus.
Abstract: Many genetic loci affect circulating lipid levels, but it remains unknown whether lifestyle factors, such as physical activity, modify these genetic effects. To identify lipid loci interacting with physical activity, we performed genome-wide analyses of circulating HDL cholesterol, LDL cholesterol, and triglyceride levels in up to 120,979 individuals of European, African, Asian, Hispanic, and Brazilian ancestry, with follow-up of suggestive associations in an additional 131,012 individuals. We find four loci, in/near CLASP1, LHX1, SNTA1, and CNTNAP2, that are associated with circulating lipid levels through interaction with physical activity; higher levels of physical activity enhance the HDL cholesterol-increasing effects of the CLASP1, LHX1, and SNTA1 loci and attenuate the LDL cholesterol-increasing effect of the CNTNAP2 locus. The CLASP1, LHX1, and SNTA1 regions harbor genes linked to muscle function and lipid metabolism. Our results elucidate the role of physical activity interactions in the genetic contribution to blood lipid levels.

Journal ArticleDOI
David M. Brazel1, Yu Jiang2, Jordan M. Hughey2, Valérie Turcot3  +182 moreInstitutions (28)
TL;DR: Fine-mapping genome-wide association study loci identifies specific variants contributing to the biological etiology of substance use behavior, including nonsynonymous/loss-of-function coding variants.

Journal ArticleDOI
Evangelos Evangelou1, Evangelos Evangelou2, He Gao2, Congying Chu3, Georgios Ntritsos1, Paul Blakeley2, Andrew R. Butts4, Raha Pazoki2, Hideaki Suzuki5, Hideaki Suzuki2, Fotios Koskeridis1, Andrianos M. Yiorkas6, Andrianos M. Yiorkas2, Ibrahim Karaman2, Joshua Elliott2, Qiang Luo7, Qiang Luo8, Stefanie Aeschbacher9, Traci M. Bartz10, Sebastian E. Baumeister11, Sebastian E. Baumeister12, Peter S. Braund13, Peter S. Braund14, Michael R. Brown15, Jennifer A. Brody10, Toni-Kim Clarke16, Niki Dimou1, Jessica D. Faul17, Georg Homuth12, Anne U. Jackson17, Katherine A. Kentistou16, Peter K. Joshi16, Rozenn N. Lemaitre10, Penelope A. Lind18, Leo-Pekka Lyytikäinen, Massimo Mangino19, Massimo Mangino3, Yuri Milaneschi20, Christopher P. Nelson13, Christopher P. Nelson14, Ilja M. Nolte21, Mia-Maria Perälä22, Ozren Polasek23, David J. Porteous16, Scott M. Ratliff17, Jennifer A. Smith17, Alena Stančáková24, Alexander Teumer25, Samuli Tuominen26, Sébastien Thériault27, Sébastien Thériault28, Jagadish Vangipurapu24, John Whitfield18, Alexis C. Wood29, Jie Yao30, Bing Yu15, Wei Zhao17, Dan E. Arking31, Juha Auvinen32, Chunyu Liu33, Minna Männikkö32, Lorenz Risch34, Jerome I. Rotter35, Harold Snieder21, Juha Veijola32, Alexandra I. F. Blakemore6, Alexandra I. F. Blakemore2, Michael Boehnke17, Harry Campbell16, David Conen28, Johan G. Eriksson22, Johan G. Eriksson26, Hans J. Grabe25, Hans J. Grabe36, Xiuqing Guo30, Pim van der Harst21, Catharina A. Hartman21, Caroline Hayward16, Andrew C. Heath37, Marjo-Riitta Järvelin32, Marjo-Riitta Järvelin6, Marjo-Riitta Järvelin2, Mika Kähönen38, Sharon L.R. Kardia17, Michael Kühne9, Johanna Kuusisto24, Markku Laakso24, Jari Lahti26, Terho Lehtimäki, Andrew M. McIntosh16, Karen L. Mohlke39, Alanna C. Morrison15, Nicholas G. Martin18, Albertine J. Oldehinkel21, Brenda W.J.H. Penninx20, Bruce M. Psaty40, Bruce M. Psaty10, Olli T. Raitakari41, Igor Rudan16, Nilesh J. Samani14, Nilesh J. Samani13, Laura J. Scott17, Tim D. Spector3, Niek Verweij21, David R. Weir17, James F. Wilson16, Daniel Levy42, Ioanna Tzoulaki2, Jimmy D. Bell43, Paul M. Matthews2, Adrian Rothenfluh4, Sylvane Desrivières3, Gunter Schumann3, Gunter Schumann44, Paul Elliott 
TL;DR: This article conducted a meta-analysis of genome-wide association studies of alcohol consumption from the UK Biobank, the Alcohol Genome Wide Consortium and the Cohorts for Heart and Aging Research in Genomic Epidemiology Plus consortia, collecting data from 480,842 people of European descent to decipher the genetic architecture of alcohol intake.
Abstract: Excessive alcohol consumption is one of the main causes of death and disability worldwide. Alcohol consumption is a heritable complex trait. Here we conducted a meta-analysis of genome-wide association studies of alcohol consumption (g d-1) from the UK Biobank, the Alcohol Genome-Wide Consortium and the Cohorts for Heart and Aging Research in Genomic Epidemiology Plus consortia, collecting data from 480,842 people of European descent to decipher the genetic architecture of alcohol intake. We identified 46 new common loci and investigated their potential functional importance using magnetic resonance imaging data and gene expression studies. We identify genetic pathways associated with alcohol consumption and suggest genetic mechanisms that are shared with neuropsychiatric disorders such as schizophrenia.

Journal ArticleDOI
TL;DR: This study establishes MYRF as a nanophthalmos gene and uncovers a new pathway for eye growth and development.
Abstract: Nanophthalmos is a rare, potentially devastating eye condition characterized by small eyes with relatively normal anatomy, a high hyperopic refractive error, and frequent association with angle closure glaucoma and vision loss. The condition constitutes the extreme of hyperopia or farsightedness, a common refractive error that is associated with strabismus and amblyopia in children. NNO1 was the first mapped nanophthalmos locus. We used combined pooled exome sequencing and strong linkage data in the large family used to map this locus to identify a canonical splice site alteration upstream of the last exon of the gene encoding myelin regulatory factor (MYRF c.3376-1G>A), a membrane bound transcription factor that undergoes autoproteolytic cleavage for nuclear localization. This variant produced a stable RNA transcript, leading to a frameshift mutation p.Gly1126Valfs*31 in the C-terminus of the protein. In addition, we identified an early truncating MYRF frameshift mutation, c.769dupC (p.S264QfsX74), in a patient with extreme axial hyperopia and syndromic features. Myrf conditional knockout mice (CKO) developed depigmentation of the retinal pigment epithelium (RPE) and retinal degeneration supporting a role of this gene in retinal and RPE development. Furthermore, we demonstrated the reduced expression of Tmem98, another known nanophthalmos gene, in Myrf CKO mice, and the physical interaction of MYRF with TMEM98. Our study establishes MYRF as a nanophthalmos gene and uncovers a new pathway for eye growth and development.

Evangelos Evangelou, He Gao, Congying Chu, Georgios Ntritsos, Paul Blakeley, Andrew R. Butts, Raha Pazoki, Hideaki Suzuki, Fotios Koskeridis, Andrianos M. Yiorkas, Ibrahim Karaman, Joshua Elliott, Qiang Luo, Stefanie Aeschbacher, Traci M. Bartz, Sebastian E. Baumeister, Peter S. Braund, Michael R. Brown, Jennifer A. Brody, Toni-Kim Clarke, Niki Dimou, Jessica D. Faul, Georg Homuth, Anne U. Jackson, Katherine A. Kentistou, Peter K. Joshi, Rozenn N. Lemaitre, Penelope A. Lind, Leo-Pekka Lyytikäinen, Massimo Mangino, Yuri Milaneschi, Christopher P. Nelson, Ilja M. Nolte, Mia-Maria Perälä, Ozren Polasek, David J. Porteous, Scott M. Ratliff, Jennifer A. Smith, Alena Stančáková, Alexander Teumer, Samuli Tuominen, Sébastien Thériault, Jagadish Vangipurapu, John Whitfield, Alexis C. Wood, Jie Yao, Bing Yu, Wei Zhao, Dan E. Arking, Juha Auvinen, Chunyu Liu, Minna Männikkö, Lorenz Risch, Rotter, Jerome, I, Harold Snieder, Juha Veijola, Blakemore, Alexandra, I, Michael Boehnke, Harry Campbell, David Conen, Johan G. Eriksson, Hans J. Grabe, Xiuqing Guo, Pim van der Harst, Catharina A. Hartman, Caroline Hayward, Andrew Heath, Marjo-Riitta Järvelin, Mika Kähönen, Sharon L.R. Kardia, Michael Kühne, Johanna Kuusisto, Maru Laakso, Jari Lahti, Terho Lehtimäki, Andrew M. McIntosh, Karen L. Mohlke, Alanna C. Morrison, Nicholas G. Martin, Albertine J. Oldehinkel, Brenda W.J.H. Penninx, Bruce M. Psaty, T. Raitakari, Igor Rudan, Nilesh J. Samani, Laura J. Scott, Tim D. Spector, Niek Verweij, David R. Weir, James F. Wilson, Daniel Levy, Ioanna Tzoulaki, Jimmy D. Bell, Paul M. Matthews, Adrian Rothenfluh, Sylvane Desrivières, Gunter Schumann, Paul Elliott 
01 Jan 2019
TL;DR: A meta-analysis of genome-wide association studies of alcohol consumption from the UK Biobank, the Alcohol Genome-Wide Consortium and the Cohorts for Heart and Aging Research in Genomic Epidemiology Plus consortia identifies 46 new common loci associated with alcohol consumption and suggests genetic mechanisms that are shared with neuropsychiatric disorders such as schizophrenia.

Journal ArticleDOI
TL;DR: A general framework for testing pleiotropic effects of rare variants on multiple continuous phenotypes using multivariate kernel regression (Multi‐SKAT), which can improve power over single‐phenotype SKAT‐O test and existing multiple‐phenotypes tests, while maintaining Type I error rate.
Abstract: In genetic association analysis, a joint test of multiple distinct phenotypes can increase power to identify sets of trait-associated variants within genes or regions of interest. Existing multiphenotype tests for rare variants make specific assumptions about the patterns of association with underlying causal variants, and the violation of these assumptions can reduce power to detect association. Here, we develop a general framework for testing pleiotropic effects of rare variants on multiple continuous phenotypes using multivariate kernel regression (Multi-SKAT). Multi-SKAT models affect sizes of variants on the phenotypes through a kernel matrix and perform a variance component test of association. We show that many existing tests are equivalent to specific choices of kernel matrices with the Multi-SKAT framework. To increase power of detecting association across tests with different kernel matrices, we developed a fast and accurate approximation of the significance of the minimum observed P value across tests. To account for related individuals, our framework uses random effects for the kinship matrix. Using simulated data and amino acid and exome-array data from the METabolic Syndrome In Men (METSIM) study, we show that Multi-SKAT can improve power over single-phenotype SKAT-O test and existing multiple-phenotype tests, while maintaining Type I error rate.

01 Jan 2019
TL;DR: It is found that physical activity modifies the effects of four genetic loci on HDL or LDL cholesterol, and higher levels of physical activity enhance the HDL cholesterol-increasing effects of the CLASP1, LHX1, and SNTA1 loci and attenuate the LDL cholesterol- Increasing effect of the CNTNAP2 locus.
Abstract: Many genetic loci affect circulating lipid levels, but it remains unknown whether lifestyle factors, such as physical activity, modify these genetic effects. To identify lipid loci interacting with physical activity, we performed genome-wide analyses of circulating HDL cholesterol, LDL cholesterol, and triglyceride levels in up to 120,979 individuals of European, African, Asian, Hispanic, and Brazilian ancestry, with follow-up of suggestive associations in an additional 131,012 individuals. We find four loci, in/near CLASP1, LHX1, SNTA1, and CNTNAP2, that are associated with circulating lipid levels through interaction with physical activity; higher levels of physical activity enhance the HDL cholesterol-increasing effects of the CLASP1, LHX1, and SNTA1 loci and attenuate the LDL cholesterol-increasing effect of the CNTNAP2 locus. The CLASP1, LHX1, and SNTA1 regions harbor genes linked to muscle function and lipid metabolism. Our results elucidate the role of physical activity interactions in the genetic contribution to blood lipid levels.GWAS have identified more than 500 genetic loci associated with blood lipid levels. Here, the authors report a genome-wide analysis of interactions between genetic markers and physical activity, and find that physical activity modifies the effects of four genetic loci on HDL or LDL cholesterol.

Journal ArticleDOI
TL;DR: Evidence of colocalization is reevaluated using two approaches, conditional analysis and the Bayesian test COLOC, and it is shown that providing COLOC with approximate conditional summary statistics at multi-signal GWAS loci can reconcile disagreements in colocalized classification between the two tests.
Abstract: Integration of genome-wide association study (GWAS) signals with expression quantitative trait loci (eQTL) studies enables identification of candidate genes. However, evaluating whether nearby signals may share causal variants, termed colocalization, is affected by the presence of allelic heterogeneity, different variants at the same locus impacting the same phenotype. We previously identified eQTL in subcutaneous adipose tissue from 770 participants in the Metabolic Syndrome in Men (METSIM) study and detected 15 eQTL signals that colocalized with GWAS signals for waist-hip ratio adjusted for body mass index (WHRadjBMI) from the Genetic Investigation of Anthropometric Traits consortium. Here, we reevaluated evidence of colocalization using two approaches, conditional analysis and the Bayesian test COLOC, and show that providing COLOC with approximate conditional summary statistics at multi-signal GWAS loci can reconcile disagreements in colocalization classification between the two tests. Next, we performed conditional analysis on the METSIM subcutaneous adipose tissue data to identify conditionally distinct or secondary eQTL signals. We used the two approaches to test for colocalization with WHRadjBMI GWAS signals and evaluated the differences in colocalization classification between the two tests. Through these analyses, we identified four GWAS signals colocalized with secondary eQTL signals for FAM13A, SSR3, GRB14 and FMO1. Thus, at loci with multiple eQTL and/or GWAS signals, analyzing each signal independently enabled additional candidate genes to be identified.

Journal ArticleDOI
TL;DR: This work used subcutaneous adipose tissue RNA-seq data from 434 Finnish men from the METSIM study to identify 9,687 primary and 2,785 secondary cis-expression quantitative trait loci (eQTL), identifying hundreds of candidate genes that may act in adipose tissues to influence cardiometabolic traits.
Abstract: Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with cardiometabolic traits including type 2 diabetes (T2D), lipid levels, body fat distribution, and adiposity, although most causal genes remain unknown. We used subcutaneous adipose tissue RNA-seq data from 434 Finnish men from the METSIM study to identify 9,687 primary and 2,785 secondary cis-expression quantitative trait loci (eQTL;

Journal ArticleDOI
Yun Ju Sung1, Lisa de las Fuentes1, Thomas W. Winkler2, Daniel I. Chasman3  +314 moreInstitutions (101)
TL;DR: A genome-wide gene-smoking interaction study of mean arterial pressure (MAP) and pulse pressure (PP) in 129 913 individuals in stage 1 and follow-up analysis in 480 178 additional individuals in stages 2 identified 136 loci significantly associated with MAP and/or PP and identified nine new signals near known loci.
Abstract: Elevated blood pressure (BP), a leading cause of global morbidity and mortality, is influenced by both genetic and lifestyle factors. Cigarette smoking is one such lifestyle factor. Across five ancestries, we performed a genome-wide gene-smoking interaction study of mean arterial pressure (MAP) and pulse pressure (PP) in 129 913 individuals in stage 1 and follow-up analysis in 480 178 additional individuals in stage 2. We report here 136 loci significantly associated with MAP and/or PP. Of these, 61 were previously published through main-effect analysis of BP traits, 37 were recently reported by us for systolic BP and/or diastolic BP through gene-smoking interaction analysis and 38 were newly identified (P < 5 × 10-8, false discovery rate < 0.05). We also identified nine new signals near known loci. Of the 136 loci, 8 showed significant interaction with smoking status. They include CSMD1 previously reported for insulin resistance and BP in the spontaneously hypertensive rats. Many of the 38 new loci show biologic plausibility for a role in BP regulation. SLC26A7 encodes a chloride/bicarbonate exchanger expressed in the renal outer medullary collecting duct. AVPR1A is widely expressed, including in vascular smooth muscle cells, kidney, myocardium and brain. FHAD1 is a long non-coding RNA overexpressed in heart failure. TMEM51 was associated with contractile function in cardiomyocytes. CASP9 plays a central role in cardiomyocyte apoptosis. Identified only in African ancestry were 30 novel loci. Our findings highlight the value of multi-ancestry investigations, particularly in studies of interaction with lifestyle factors, where genomic and lifestyle differences may contribute to novel findings.

01 Jan 2019
TL;DR: This study integrates genetic, diverse -omics, and physiological measurements—a challenge of increasing importance in the field of human genomics—and identifies hundreds of genes and thousands of DNAme sites that may drive the reported disease/quantitative trait genetic associations.
Abstract: We integrate comeasured gene expression and DNA methylation (DNAme) in 265 human skeletal muscle biopsies from the FUSION study with >7 million genetic variants and eight physiological traits: height, waist, weight, waist-hip ratio, body mass index, fasting serum insulin, fasting plasma glucose, and type 2 diabetes. We find hundreds of genes and DNAme sites associated with fasting insulin, waist, and body mass index, as well as thousands of DNAme sites associated with gene expression (eQTM). We find that controlling for heterogeneity in tissue/muscle fiber type reduces the number of physiological trait associations, and that long-range eQTMs (>1 Mb) are reduced when controlling for tissue/muscle fiber type or latent factors. We map genetic regulators (quantitative trait loci; QTLs) of expression (eQTLs) and DNAme (mQTLs). Using Mendelian randomization (MR) and mediation techniques, we leverage these genetic maps to predict 213 causal relationships between expression and DNAme, approximately two-thirds of which predict methylation to causally influence expression. We use MR to integrate FUSION mQTLs, FUSION eQTLs, and GTEx eQTLs for 48 tissues with genetic associations for 534 diseases and quantitative traits. We identify hundreds of genes and thousands of DNAme sites that may drive the reported disease/quantitative trait genetic associations. We identify 300 gene expression MR associations that are present in both FUSION and GTEx skeletal muscle and that show stronger evidence of MR association in skeletal muscle than other tissues, which may partially reflect differences in power across tissues. As one example, we find that increased RXRA muscle expression may decrease lean tissue mass.

01 Jan 2019
TL;DR: In this article, the authors used genomic inbreeding coefficients (F ROH) for >1.4 million individuals and found that F ROH is significantly associated with apparently deleterious changes in 32 out of 100 traits analysed.
Abstract: In many species, the offspring of related parents suffer reduced reproductive success, a phenomenon known as inbreeding depression. In humans, the importance of this effect has remained unclear, partly because reproduction between close relatives is both rare and frequently associated with confounding social factors. Here, using genomic inbreeding coefficients (F ROH) for >1.4 million individuals, we show that F ROH is significantly associated (p < 0.0005) with apparently deleterious changes in 32 out of 100 traits analysed. These changes are associated with runs of homozygosity (ROH), but not with common variant homozygosity, suggesting that genetic variants associated with inbreeding depression are predominantly rare. The effect on fertility is striking: F ROH equivalent to the offspring of first cousins is associated with a 55% decrease [95% CI 44–66%] in the odds of having children. Finally, the effects of F ROH are confirmed within full-sibling pairs, where the variation in F ROH is independent of all environmental confounding. Inbreeding depression has been observed in many different species, but in humans a systematic analysis has been difficult so far. Here, analysing more than 1.3 million individuals, the authors show that a genomic inbreeding coefficient (FROH) is associated with disadvantageous outcomes in 32 out of 100 traits tested.

01 Jan 2019
TL;DR: A person's lipid profile is influenced by genetic variants and alcohol consumption, but the contribution of interactions between these exposures has not been studied, so gene-alcohol interactions are incorporated into a multiancestry genome-wide association study of levels of high-density lipoprotein cholesterol, low-density cholesterol, and triglycerides.
Abstract: A person's lipid profile is influenced by genetic variants and alcohol consumption, but the contribution of interactions between these exposures has not been studied. We therefore incorporated gene-alcohol interactions into a multiancestry genome-wide association study of levels of high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and triglycerides. We included 45 studies in stage 1 (genome-wide discovery) and 66 studies in stage 2 (focused follow-up), for a total of 394,584 individuals from 5 ancestry groups. Analyses covered the period July 2014-November 2017. Genetic main effects and interaction effects were jointly assessed by means of a 2-degrees-of-freedom (df) test, and a 1-df test was used to assess the interaction effects alone. Variants at 495 loci were at least suggestively associated (P < 1 × 10-6) with lipid levels in stage 1 and were evaluated in stage 2, followed by combined analyses of stage 1 and stage 2. In the combined analysis of stages 1 and 2, a total of 147 independent loci were associated with lipid levels at P < 5 × 10-8 using 2-df tests, of which 18 were novel. No genome-wide-significant associations were found testing the interaction effect alone. The novel loci included several genes (proprotein convertase subtilisin/kexin type 5 (PCSK5), vascular endothelial growth factor B (VEGFB), and apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (APOBEC1) complementation factor (A1CF)) that have a putative role in lipid metabolism on the basis of existing evidence from cellular and experimental models.

01 Jan 2019
TL;DR: This paper performed a genome-wide gene-smoking interaction study of mean arterial pressure (MAP) and pulse pressure (PP) in 129, 913 individuals in stage 1 and follow-up analysis in 480,178 additional individuals in stages 2.
Abstract: Elevated blood pressure (BP), a leading cause of global morbidity and mortality, is influenced by both genetic and lifestyle factors. Cigarette smoking is one such lifestyle factor. Across five ancestries, we performed a genome-wide gene-smoking interaction study of mean arterial pressure (MAP) and pulse pressure (PP) in 129 913 individuals in stage 1 and follow-up analysis in 480 178 additional individuals in stage 2. We report here 136 loci significantly associated with MAP and/or PP. Of these, 61 were previously published through main-effect analysis of BP traits, 37 were recently reported by us for systolic BP and/or diastolic BP through gene-smoking interaction analysis and 38 were newly identified (P < 5 × 10-8, false discovery rate < 0.05). We also identified nine new signals near known loci. Of the 136 loci, 8 showed significant interaction with smoking status. They include CSMD1 previously reported for insulin resistance and BP in the spontaneously hypertensive rats. Many of the 38 new loci show biologic plausibility for a role in BP regulation. SLC26A7 encodes a chloride/bicarbonate exchanger expressed in the renal outer medullary collecting duct. AVPR1A is widely expressed, including in vascular smooth muscle cells, kidney, myocardium and brain. FHAD1 is a long non-coding RNA overexpressed in heart failure. TMEM51 was associated with contractile function in cardiomyocytes. CASP9 plays a central role in cardiomyocyte apoptosis. Identified only in African ancestry were 30 novel loci. Our findings highlight the value of multi-ancestry investigations, particularly in studies of interaction with lifestyle factors, where genomic and lifestyle differences may contribute to novel findings.

Posted ContentDOI
31 May 2019-bioRxiv
TL;DR: These findings illustrate the advantages of performing functional and regulatory studies in tissues of greatest disease-relevance while expanding the mechanistic insights into complex traits association loci activity with an expanded list of putative transcripts implicated in T2D development.
Abstract: Most signals detected by genome-wide association studies map to non-coding sequence and their tissue-specific effects influence transcriptional regulation. However, many key tissues and cell-types required for appropriate functional inference are absent from large-scale resources such as ENCODE and GTEx. We explored the relationship between genetic variants influencing predisposition to type 2 diabetes (T2D) and related glycemic traits, and human pancreatic islet transcription using RNA-Seq and genotyping data from 420 islet donors. We find: (a) eQTLs have a variable replication rate across the 44 GTEx tissues (<73%), indicating that our study captured islet-specific cis-eQTL signals; (b) islet eQTL signals show marked overlap with islet epigenome annotation, though eQTL effect size is reduced in the stretch enhancers most strongly implicated in GWAS signal location; (c) selective enrichment of islet eQTL overlap with the subset of T2D variants implicated in islet dysfunction; and (d) colocalization between islet eQTLs and variants influencing T2D or related glycemic traits, delivering candidate effector transcripts at 23 loci, including DGKB and TCF7L2. Our findings illustrate the advantages of performing functional and regulatory studies in tissues of greatest disease-relevance while expanding our mechanistic insights into complex traits association loci activity with an expanded list of putative transcripts implicated in T2D development.

Journal ArticleDOI
Cassandra N. Spracklen1, Tugce Karaderi, Hanieh Yaghootkar2, Hanieh Yaghootkar3, Claudia Schurmann4, Rebecca S. Fine5, Rebecca S. Fine6, Rebecca S. Fine7, Zoltán Kutalik2, Zoltán Kutalik8, Zoltán Kutalik9, Michael Preuss4, Yingchang Lu4, Yingchang Lu10, Laura B. L. Wittemans11, Laura B. L. Wittemans12, Linda S. Adair1, Matthew A. Allison13, Najaf Amin14, Paul L. Auer15, Traci M. Bartz16, Matthias Blüher17, Michael Boehnke18, Judith B. Borja19, Jette Bork-Jensen20, Linda Broer21, Daniel I. Chasman22, Daniel I. Chasman7, Yii-Der Ida Chen23, Paraskevi Chirstofidou24, Ayse Demirkan14, Cornelia M. van Duijn14, Mary F. Feitosa25, Melissa E. Garcia26, Mariaelisa Graff1, Harald Grallert, Niels Grarup20, Xiuqing Guo23, Jeffrey Haesser27, Torben Hansen20, Tamara B. Harris26, Heather M. Highland1, Jaeyoung Hong28, M. Arfan Ikram21, Erik Ingelsson29, Erik Ingelsson30, Rebecca D. Jackson31, Pekka Jousilahti26, Mika Kähönen32, Jorge R. Kizer33, Peter Kovacs17, Jennifer Kriebel, Markku Laakso34, Leslie A. Lange35, Terho Lehtimäki, Jin Li29, Ruifang Li-Gao36, Lars Lind30, Jian'an Luan12, Leo-Pekka Lyytikäinen, Stuart MacGregor37, David A. Mackey38, Anubha Mahajan11, Anubha Mahajan39, Massimo Mangino40, Massimo Mangino24, Satu Männistö26, Mark I. McCarthy11, Mark I. McCarthy39, Barbara McKnight16, Carolina Medina-Gomez21, James B. Meigs6, James B. Meigs7, Sophie Molnos, Dennis O. Mook-Kanamori36, Andrew P. Morris41, Andrew P. Morris11, Renée de Mutsert36, Mike A. Nalls26, Ivana Nedeljkovic14, Kari E. North1, Craig E. Pennell42, Aruna D. Pradhan7, Aruna D. Pradhan22, Michael A. Province25, Olli T. Raitakari43, Olli T. Raitakari44, Chelsea K. Raulerson1, Alex P. Reiner27, Paul M. Ridker7, Paul M. Ridker22, Samuli Ripatti6, Samuli Ripatti45, Neil Roberston11, Neil Roberston39, Jerome I. Rotter23, Veikko Salomaa26, America A. Sandoval-Zárate, Colleen M. Sitlani16, Tim D. Spector24, Konstantin Strauch46, Michael Stumvoll17, Kent D. Taylor23, Betina H. Thuesen47, Anke Tönjes17, André G. Uitterlinden21, Cristina Venturini24, Mark Walker48, Carol A. Wang42, Shuai Wang28, Nicholas J. Wareham12, Sara M. Willems12, Ko Willems van Dijk36, James G. Wilson49, Ying Wu1, Jie Yao23, Kristin L. Young1, Claudia Langenberg12, Timothy M. Frayling2, Tuomas O. Kilpeläinen4, Tuomas O. Kilpeläinen20, Cecilia M. Lindgren11, Cecilia M. Lindgren6, Cecilia M. Lindgren39, Ruth J. F. Loos4, Karen L. Mohlke1 
University of North Carolina at Chapel Hill1, Royal Devon and Exeter Hospital2, University of Westminster3, Icahn School of Medicine at Mount Sinai4, Boston Children's Hospital5, Broad Institute6, Harvard University7, University of Lausanne8, Swiss Institute of Bioinformatics9, Vanderbilt University10, Wellcome Trust Centre for Human Genetics11, University of Cambridge12, University of California, San Diego13, Erasmus University Medical Center14, University of Wisconsin–Milwaukee15, University of Washington16, Leipzig University17, University of Michigan18, University of San Carlos19, University of Copenhagen20, Erasmus University Rotterdam21, Brigham and Women's Hospital22, UCLA Medical Center23, King's College London24, Washington University in St. Louis25, National Institutes of Health26, Fred Hutchinson Cancer Research Center27, Boston University28, Stanford University29, Uppsala University30, Ohio State University31, University of Tampere32, Albert Einstein College of Medicine33, University of Eastern Finland34, University of Colorado Denver35, Leiden University Medical Center36, QIMR Berghofer Medical Research Institute37, University of Western Australia38, University of Oxford39, Guy's and St Thomas' NHS Foundation Trust40, University of Liverpool41, University of Newcastle42, University of Turku43, Turku University Hospital44, University of Helsinki45, Ludwig Maximilian University of Munich46, Frederiksberg Hospital47, Newcastle University48, University of Mississippi Medical Center49
TL;DR: An exome array meta-analysis of 265,780 genetic variants in 67,739 individuals of European, Hispanic, African American, and East Asian ancestry identified 20 loci associated with adiponectin, including 11 that had been reported previously and eight of the nine loci were associated with at least one obesity or lipid trait.
Abstract: Circulating levels of adiponectin, an adipocyte-secreted protein associated with cardiovascular and metabolic risk, are highly heritable. To gain insights into the biology that regulates adiponectin levels, we performed an exome array meta-analysis of 265,780 genetic variants in 67,739 individuals of European, Hispanic, African American, and East Asian ancestry. We identified 20 loci associated with adiponectin, including 11 that had been reported previously (p .60) spanning as much as 900 kb. To identify potential genes and mechanisms through which the previously unreported association signals act to affect adiponectin levels, we assessed cross-trait associations, expression quantitative trait loci in subcutaneous adipose, and biological pathways of nearby genes. Eight of the nine loci were also associated (p

01 Jan 2019
TL;DR: A transancestral exome-wide association study for body-fat distribution identifies protein-coding variants that are significantly associated with waist-to-hip ratio adjusted for body mass index.

Journal ArticleDOI
TL;DR: In this article, sexually dimorphic associations between the carbamoyl-phosphate synthase 1 locus and the metabolic pathway leading from choline to urea were found.
Abstract: Background Recent studies have revealed sexually dimorphic associations between the carbamoyl‐phosphate synthase 1 locus, intermediates of the metabolic pathway leading from choline to urea, and ri...

Posted ContentDOI
Nicola Whiffin1, Nicola Whiffin2, Nicola Whiffin3, Konrad J. Karczewski3  +190 moreInstitutions (22)
07 Feb 2019-bioRxiv
TL;DR: In this article, the authors describe a systematic genome-wide study of variants that create and disrupt human uORFs, and explore their role in human disease using 15,708 whole genome sequences collected by the Genome Aggregation Database (gnomAD) project.
Abstract: Upstream open reading frames (uORFs) are important tissue-specific cis-regulators of protein translation. Although isolated case reports have shown that variants that create or disrupt uORFs can cause disease, genetic sequencing approaches typically focus on protein-coding regions and ignore these variants. Here, we describe a systematic genome-wide study of variants that create and disrupt human uORFs, and explore their role in human disease using 15,708 whole genome sequences collected by the Genome Aggregation Database (gnomAD) project. We show that 14,897 variants that create new start codons upstream of the canonical coding sequence (CDS), and 2,406 variants disrupting the stop site of existing uORFs, are under strong negative selection. Furthermore, variants creating uORFs that overlap the CDS show signals of selection equivalent to coding loss-of-function variants, and uORF-perturbing variants are under strong selection when arising upstream of known disease genes and genes intolerant to loss-of-function variants. Finally, we identify specific genes where perturbation of uORFs is likely to represent an important disease mechanism, and report a novel uORF frameshift variant upstream of NF2 in families with neurofibromatosis. Our results highlight uORF-perturbing variants as an important and under-recognised functional class that can contribute to penetrant human disease, and demonstrate the power of large-scale population sequencing data to study the deleteriousness of specific classes of non-coding variants.