scispace - formally typeset
Search or ask a question

Showing papers by "Daniel I. Chasman published in 2016"


Journal ArticleDOI
TL;DR: Among participants at high genetic risk, a favorable lifestyle was associated with a nearly 50% lower relative risk of coronary artery disease than was an unfavorable lifestyle, and across four studies involving 55,685 participants, genetic and lifestyle factors were independently associated with susceptibility to coronary arteries disease.
Abstract: BackgroundBoth genetic and lifestyle factors contribute to individual-level risk of coronary artery disease. The extent to which increased genetic risk can be offset by a healthy lifestyle is unknown. MethodsUsing a polygenic score of DNA sequence polymorphisms, we quantified genetic risk for coronary artery disease in three prospective cohorts — 7814 participants in the Atherosclerosis Risk in Communities (ARIC) study, 21,222 in the Women’s Genome Health Study (WGHS), and 22,389 in the Malmo Diet and Cancer Study (MDCS) — and in 4260 participants in the cross-sectional BioImage Study for whom genotype and covariate data were available. We also determined adherence to a healthy lifestyle among the participants using a scoring system consisting of four factors: no current smoking, no obesity, regular physical activity, and a healthy diet. ResultsThe relative risk of incident coronary events was 91% higher among participants at high genetic risk (top quintile of polygenic scores) than among those at low gen...

915 citations


Journal ArticleDOI
Padhraig Gormley, Verneri Anttila1, Verneri Anttila2, Bendik S. Winsvold3, Bendik S. Winsvold4, Priit Palta5, Tõnu Esko6, Tõnu Esko7, Tõnu Esko1, Tune H. Pers, Kai-How Farh2, Kai-How Farh8, Kai-How Farh1, Ester Cuenca-León, Mikko Muona, Nicholas A. Furlotte, Tobias Kurth9, Tobias Kurth10, Andres Ingason11, George McMahon12, Lannie Ligthart13, Gisela M. Terwindt14, Mikko Kallela15, Tobias Freilinger16, Tobias Freilinger17, Caroline Ran18, Scott G. Gordon19, Anine H. Stam14, Stacy Steinberg11, Guntram Borck20, Markku Koiranen21, Lydia Quaye22, Hieab H.H. Adams23, Terho Lehtimäki24, Antti-Pekka Sarin5, Juho Wedenoja5, David A. Hinds, Julie E. Buring9, Julie E. Buring2, Markus Schürks25, Paul M. Ridker9, Paul M. Ridker2, Maria Gudlaug Hrafnsdottir, Hreinn Stefansson11, Susan M. Ring12, Jouke-Jan Hottenga13, Brenda W.J.H. Penninx13, Markus Färkkilä15, Ville Artto15, Mari A. Kaunisto5, Salli Vepsäläinen15, Rainer Malik16, Andrew C. Heath26, Pamela A. F. Madden26, Nicholas G. Martin19, Grant W. Montgomery19, Mitja I. Kurki, Mart Kals7, Reedik Mägi7, Kalle Pärn7, Eija Hamalainen5, Hailiang Huang2, Hailiang Huang1, Andrea Byrnes1, Andrea Byrnes2, Lude Franke27, Jie Huang28, Evie Stergiakouli12, Phil Lee2, Phil Lee1, Cynthia Sandor29, Caleb Webber29, Zameel M. Cader29, Zameel M. Cader30, Bertram Müller-Myhsok31, Stefan Schreiber32, Thomas Meitinger33, Johan G. Eriksson34, Johan G. Eriksson5, Veikko Salomaa34, Kauko Heikkilä5, Elizabeth Loehrer23, Elizabeth Loehrer2, André G. Uitterlinden23, Albert Hofman23, Cornelia M. van Duijn23, Lynn Cherkas22, Linda M. Pedersen3, Audun Stubhaug4, Audun Stubhaug3, Christopher Sivert Nielsen3, Christopher Sivert Nielsen35, Minna Männikkö21, Evelin Mihailov7, Lili Milani7, Hartmut Göbel, Ann-Louise Esserlind36, Anne Francke Christensen36, Thomas Hansen36, Thomas Werge37, Thomas Werge38, Thomas Werge36, Jaakko Kaprio5, Jaakko Kaprio34, Arpo Aromaa34, Olli T. Raitakari39, Olli T. Raitakari40, M. Arfan Ikram23, Tim D. Spector22, Marjo-Riitta Järvelin, Andres Metspalu7, Christian Kubisch41, David P. Strachan42, Michel D. Ferrari14, Andrea Carmine Belin18, Martin Dichgans16, Maija Wessman5, Arn M. J. M. van den Maagdenberg14, John-Anker Zwart4, John-Anker Zwart3, Dorret I. Boomsma13, George Davey Smith12, Kari Stefansson11, Kari Stefansson43, Nicholas Eriksson, Mark J. Daly2, Mark J. Daly1, Benjamin M. Neale1, Benjamin M. Neale2, Jes Olesen36, Daniel I. Chasman9, Daniel I. Chasman2, Dale R. Nyholt44, Aarno Palotie 
TL;DR: For example, the authors identified 44 independent single-nucleotide polymorphisms (SNPs) significantly associated with migraine risk (P < 5 × 10−8) that mapped to 38 distinct genomic loci, including 28 loci not previously reported and a locus that to date is the first to be identified on chromosome X.
Abstract: Migraine is a debilitating neurological disorder affecting around one in seven people worldwide, but its molecular mechanisms remain poorly understood. There is some debate about whether migraine is a disease of vascular dysfunction or a result of neuronal dysfunction with secondary vascular changes. Genome-wide association (GWA) studies have thus far identified 13 independent loci associated with migraine. To identify new susceptibility loci, we carried out a genetic study of migraine on 59,674 affected subjects and 316,078 controls from 22 GWA studies. We identified 44 independent single-nucleotide polymorphisms (SNPs) significantly associated with migraine risk (P < 5 × 10−8) that mapped to 38 distinct genomic loci, including 28 loci not previously reported and a locus that to our knowledge is the first to be identified on chromosome X. In subsequent computational analyses, the identified loci showed enrichment for genes expressed in vascular and smooth muscle tissues, consistent with a predominant theory of migraine that highlights vascular etiologies.

471 citations


Journal ArticleDOI
Cristian Pattaro, Alexander Teumer1, Mathias Gorski2, Audrey Y. Chu3  +732 moreInstitutions (157)
TL;DR: A meta-analysis of genome-wide association studies for estimated glomerular filtration rate suggests that genetic determinants of eGFR are mediated largely through direct effects within the kidney and highlight important cell types and biological pathways.
Abstract: Reduced glomerular filtration rate defines chronic kidney disease and is associated with cardiovascular and all-cause mortality. We conducted a meta-analysis of genome-wide association studies for estimated glomerular filtration rate (eGFR), combining data across 133,413 individuals with replication in up to 42,166 individuals. We identify 24 new and confirm 29 previously identified loci. Of these 53 loci, 19 associate with eGFR among individuals with diabetes. Using bioinformatics, we show that identified genes at eGFR loci are enriched for expression in kidney tissues and in pathways relevant for kidney development and transmembrane transporter activity, kidney structure, and regulation of glucose metabolism. Chromatin state mapping and DNase I hypersensitivity analyses across adult tissues demonstrate preferential mapping of associated variants to regulatory regions in kidney but not extra-renal tissues. These findings suggest that genetic determinants of eGFR are mediated largely through direct effects within the kidney and highlight important cell types and biological pathways.

409 citations


Journal ArticleDOI
Nathan O. Stitziel1, Kathleen Stirrups2, Nicholas G. D. Masca3, Jeanette Erdmann, Paola G. Ferrario4, Inke R. Koenig4, Peter Weeke5, Tom R. Webb3, Paul L. Auer6, Ursula M. Schick, Yingchang Lu, He Zhang7, Marie-Pierre Dubé8, Anuj Goel9, Martin Farrall9, Gina M. Peloso10, Hong-Hee Won11, Ron Do, Erik P A Van Iperen12, Stavroula Kanoni13, Jochen Kruppa14, Anubha Mahajan9, Robert A. Scott2, Christina Willenborg, Peter S. Braund3, Julian C. van Capelleveen12, Alex S. F. Doney15, Louise A. Donnelly15, Rosanna Asselta, Piera Angelica Merlini, Stefano Duga, Nicola Marziliano16, Josh C. Denny17, Christian M. Shaffer17, Nour Eddine El-Mokhtari, Andre Franke18, Omri Gottesman, Stefanie Heilmann19, Christian Hengstenberg, Per Hoffmann20, Oddgeir L. Holmen21, Kristian Hveem, Jan-Håkan Jansson22, Karl-Heinz Joeckel23, Thorsten Kessler24, Jennifer Kriebel, Karl L. Laugwitz24, Eirini Marouli13, Nicola Martinelli16, Mark I. McCarthy9, Natalie R. van Zuydam9, Christa Meisinger, Tõnu Esko25, Evelin Mihailov25, Stefan A. Escher26, Maris Alver25, Susanne Moebus23, Andrew D. Morris, Martina Mueller-Nurasyid, Majid Nikpay27, Oliviero Olivieri16, Louis-Philippe Lemieux Perreault8, Alaa AlQarawi28, Neil R. Robertson9, Karen O. Akinsanya29, Dermot F. Reilly29, Thomas F. Vogt29, Wu Yin29, Folkert W. Asselbergs, Charles Kooperberg1, Rebecca D. Jackson30, Eli A. Stahl, Konstantin Strauch, Tibor V. Varga26, Melanie Waldenberger, Lingyao Zeng24, Aldi T. Kraja1, Chunyu Liu31, Georg Ehret32, Christopher Newton-Cheh10, Daniel I. Chasman33, Rajiv Chowdhury2, Marco M Ferrario34, Ian Ford35, J. Wouter Jukema36, Frank Kee37, Kari Kuulasmaa31, Børge G. Nordestgaard5, Markus Perola31, Danish Saleheen, Naveed Sattar35, Praveen Surendran2, David-Alexandre Trégouët38, Robin Young2, Joanna M. M. Howson2, Adam S. Butterworth2, John Danesh39, Diego Ardissino, Erwin P. Bottinger, Raimund Erbel23, Paul W. Franks26, Domenico Girelli16, Alistair S. Hall40, G. Kees Hovingh12, Adnan Kastrati24, Wolfgang Lieb18, Thomas Meitinger, William E. Kraus41, Svati H. Shah41, Ruth McPherson27, Marju Orho-Melander26, Olle Melander26, Andres Metspalu25, Colin N. A. Palmer15, Annette Peters, Daniel J. Rader42, Muredach P. Reilly42, Ruth J. F. Loos, Alexander P. Reiner1, Dan M. Roden17, Jean-Claude Tardif8, John R. Thompson3, Nicholas J. Wareham2, Hugh Watkins9, Cristen J. Willer7, Sekar Kathiresan10, Panos Deloukas28, Nilesh J. Samani3, Heribert Schunkert 
TL;DR: It was found that carriers of loss-of-function mutations in ANGPTL4 had triglyceride levels that were lower than those among noncarriers; these mutations were also associated with protection from coronary artery disease.
Abstract: BACKGROUND The discovery of low-frequency coding variants affecting the risk of coronary artery disease has facilitated the identification of therapeutic targets. METHODS Through DNA genotyping, we ...

339 citations


Journal ArticleDOI
Georg Ehret1, Georg Ehret2, Teresa Ferreira3, Daniel I. Chasman4  +372 moreInstitutions (101)
TL;DR: In this article, the authors identified 66 blood pressure-associated loci, of which 17 were new; 15 harbored multiple distinct association signals, and 66 index SNPs were enriched for cis-regulatory elements, particularly in vascular endothelial cells, consistent with a primary role in blood pressure control through modulation of vascular tone across multiple tissues.
Abstract: To dissect the genetic architecture of blood pressure and assess effects on target organ damage, we analyzed 128,272 SNPs from targeted and genome-wide arrays in 201,529 individuals of European ancestry, and genotypes from an additional 140,886 individuals were used for validation. We identified 66 blood pressure-associated loci, of which 17 were new; 15 harbored multiple distinct association signals. The 66 index SNPs were enriched for cis-regulatory elements, particularly in vascular endothelial cells, consistent with a primary role in blood pressure control through modulation of vascular tone across multiple tissues. The 66 index SNPs combined in a risk score showed comparable effects in 64,421 individuals of non-European descent. The 66-SNP blood pressure risk score was significantly associated with target organ damage in multiple tissues but with minor effects in the kidney. Our findings expand current knowledge of blood pressure-related pathways and highlight tissues beyond the classical renal system in blood pressure regulation.

332 citations


01 Jan 2016
TL;DR: The 66-SNP blood pressure risk score was significantly associated with target organ damage in multiple tissues but with minor effects in the kidney, expanding current knowledge of blood pressure–related pathways and highlighting tissues beyond the classical renal system in blood pressure regulation.

272 citations


Journal ArticleDOI
Nicola Barban1, Rick Jansen2, Ronald de Vlaming3, Ahmad Vaez4  +281 moreInstitutions (83)
TL;DR: In this article, the authors reported a large genome-wide association study of both sexes including 251,151 individuals for AB and 343,072 individuals for NEB and identified 12 independent loci that are significantly associated with AB and NEB.
Abstract: The genetic architecture of human reproductive behavior-age at first birth (AFB) and number of children ever born (NEB)-has a strong relationship with fitness, human development, infertility and risk of neuropsychiatric disorders However, very few genetic loci have been identified, and the underlying mechanisms of AFB and NEB are poorly understood We report a large genome-wide association study of both sexes including 251,151 individuals for AFB and 343,072 individuals for NEB We identified 12 independent loci that are significantly associated with AFB and/or NEB in a SNP-based genome-wide association study and 4 additional loci associated in a gene-based effort These loci harbor genes that are likely to have a role, either directly or by affecting non-local gene expression, in human reproduction and infertility, thereby increasing understanding of these complex traits

237 citations


Journal ArticleDOI
Chunyu Liu1, Aldi T. Kraja2, Jennifer A. Smith3, Jennifer A. Brody4, Nora Franceschini5, Joshua C. Bis4, Kenneth Rice4, Alanna C. Morrison6, Yingchang Lu7, Stefan Weiss8, Xiuqing Guo9, Walter Palmas10, Lisa W. Martin11, Yii-Der Ida Chen9, Praveen Surendran12, Fotios Drenos13, Fotios Drenos14, James P. Cook15, James P. Cook16, Paul L. Auer17, Audrey Y. Chu18, Ayush Giri19, Wei Zhao3, Johanna Jakobsdottir, Li-An Lin6, Jeanette M. Stafford20, Najaf Amin21, Hao Mei22, Jie Yao9, Arend Voorman23, Martin G. Larson1, Megan L. Grove6, Albert V. Smith24, Shih-Jen Hwang, Han Chen25, Tianxiao Huan, Gulum Kosova25, Gulum Kosova26, Nathan O. Stitziel2, Sekar Kathiresan25, Sekar Kathiresan26, Nilesh J. Samani27, Nilesh J. Samani16, Heribert Schunkert28, Panos Deloukas29, Panos Deloukas30, Man Li31, Christian Fuchsberger31, Cristian Pattaro32, Mathias Gorski32, Charles Kooperberg33, George J. Papanicolaou33, Jacques E. Rossouw3, Jessica D. Faul3, Sharon L.R. Kardia34, Claude Bouchard35, Leslie J. Raffel21, André G. Uitterlinden21, Oscar H. Franco1, Ramachandran S. Vasan, Christopher J. O'Donnell9, Kent D. Taylor36, Kiang Liu7, Erwin P. Bottinger7, Omri Gottesman2, E. Warwick Daw18, Franco Giulianini3, Santhi K. Ganesh37, Elias Salfati33, Tamara B. Harris33, Lenore J. Launer38, Marcus Dörr38, Stephan B. Felix8, Rainer Rettig38, Henry Völzke9, Eric Kim, Wen-Jane Lee39, Wen-Jane Lee40, I-Te Lee, Wayne H-H Sheu19, Krystal S Tsosie19, Digna R. Velez Edwards41, Yongmei Liu22, Adolfo Correa3, David R. Weir8, Uwe Völker25, Uwe Völker18, Paul M. Ridker6, Eric Boerwinkle24, Vilmundur Gudnason4, Alexander P. Reiner21, Cornelia M. van Duijn2, Ingrid B. Borecki19, Todd L. Edwards37, Aravinda Chakravarti9, Jerome I. Rotter, Bruce M. Psaty7, Ruth J. F. Loos6, Myriam Fornage37, Myriam Fornage42, Georg Ehret25, Georg Ehret26, Christopher Newton-Cheh, Daniel Levy25, Daniel Levy18, Daniel I. Chasman25 
TL;DR: This large collection of blood pressure–associated loci suggests new therapeutic strategies for hypertension, emphasizing a link with cardiometabolic risk.
Abstract: Meta-analyses of association results for blood pressure using exome-centric single-variant and gene-based tests identified 31 new loci in a discovery stage among 146,562 individuals, with follow-up and meta-analysis in 180,726 additional individuals (total n = 327,288). These blood pressure-associated loci are enriched for known variants for cardiometabolic traits. Associations were also observed for the aggregation of rare and low-frequency missense variants in three genes, NPR1, DBH, and PTPMT1. In addition, blood pressure associations at 39 previously reported loci were confirmed. The identified variants implicate biological pathways related to cardiometabolic traits, vascular function, and development. Several new variants are inferred to have roles in transcription or as hubs in protein-protein interaction networks. Genetic risk scores constructed from the identified variants were strongly associated with coronary disease and myocardial infarction. This large collection of blood pressure-associated loci suggests new therapeutic strategies for hypertension, emphasizing a link with cardiometabolic risk.

218 citations


Journal ArticleDOI
Sara L. Pulit1, Patrick F. McArdle2, Quenna Wong, Rainer Malik  +199 moreInstitutions (4)
TL;DR: A novel locus (G allele at rs12122341) at 1p13.2 near TSPAN2 that was associated with large artery atherosclerosis-related stroke was identified and supported robust associations with ischaemic stroke for four other loci that have been reported in previous studies.
Abstract: Summary Background The discovery of disease-associated loci through genome-wide association studies (GWAS) is the leading genetic approach to the identification of novel biological pathways underlying diseases in humans. Until recently, GWAS in ischaemic stroke have been limited by small sample sizes and have yielded few loci associated with ischaemic stroke. We did a large-scale GWAS to identify additional susceptibility genes for stroke and its subtypes. Methods To identify genetic loci associated with ischaemic stroke, we did a two-stage GWAS. In the first stage, we included 16 851 cases with state-of-the-art phenotyping data and 32 473 stroke-free controls. Cases were aged 16 to 104 years, recruited between 1989 and 2012, and subtypes of ischaemic stroke were recorded by centrally trained and certified investigators who used the web-based protocol, Causative Classification of Stroke (CCS). We constructed case-control strata by identifying samples that were genotyped on nearly identical arrays and were of similar genetic ancestral background. We cleaned and imputed data by use of dense imputation reference panels generated from whole-genome sequence data. We did genome-wide testing to identify stroke-associated loci within each stratum for each available phenotype, and we combined summary-level results using inverse variance-weighted fixed-effects meta-analysis. In the second stage, we did in-silico lookups of 1372 single nucleotide polymorphisms identified from the first stage GWAS in 20 941 cases and 364 736 unique stroke-free controls. The ischaemic stroke subtypes of these cases had previously been established with the Trial of Org 10 172 in Acute Stroke Treatment (TOAST) classification system, in accordance with local standards. Results from the two stages were then jointly analysed in a final meta-analysis. Findings We identified a novel locus (G allele at rs12122341) at 1p13.2 near TSPAN2 that was associated with large artery atherosclerosis-related stroke (first stage odds ratio [OR] 1·21, 95% CI 1·13–1·30, p=4·50 × 10 −8 ; joint OR 1·19, 1·12–1·26, p=1·30 × 10 −9 ). Our results also supported robust associations with ischaemic stroke for four other loci that have been reported in previous studies, including PITX2 (first stage OR 1·39, 1·29–1·49, p=3·26 × 10 −19 ; joint OR 1·37, 1·30–1·45, p=2·79 × 10 −32 ) and ZFHX3 (first stage OR 1·19, 1·11–1·27, p=2·93 × 10 −7 ; joint OR 1·17, 1·11–1·23, p=2·29 × 10 −10 ) for cardioembolic stroke, and HDAC9 (first stage OR 1·29, 1·18–1·42, p=3·50 × 10 −8 ; joint OR 1·24, 1·15–1·33, p=4·52 × 10 −9 ) for large artery atherosclerosis stroke. The 12q24 locus near ALDH2 , which has previously been associated with all ischaemic stroke but not with any specific subtype, exceeded genome-wide significance in the meta-analysis of small artery stroke (first stage OR 1·20, 1·12–1·28, p=6·82 × 10 −8 ; joint OR 1·17, 1·11–1·23, p=2·92 × 10 −9 ). Other loci associated with stroke in previous studies, including NINJ2 , were not confirmed. Interpretation Our results suggest that all ischaemic stroke-related loci previously implicated by GWAS are subtype specific. We identified a novel gene associated with large artery atherosclerosis stroke susceptibility. Follow-up studies will be necessary to establish whether the locus near TSPAN2 can be a target for a novel therapeutic approach to stroke prevention. In view of the subtype-specificity of the associations detected, the rich phenotyping data available in the Stroke Genetics Network (SiGN) are likely to be crucial for further genetic discoveries related to ischaemic stroke. Funding US National Institute of Neurological Disorders and Stroke, National Institutes of Health.

210 citations


Journal ArticleDOI
Gunter Schumann1, Chunyu Liu2, Chunyu Liu3, Paul F. O'Reilly1  +146 moreInstitutions (57)
TL;DR: It is shown that brain-specific β-Klotho KO mice have an increased alcohol preference and that FGF21 inhibits alcohol drinking by acting on the brain, suggesting that a liver–brain endocrine axis may play an important role in the regulation of alcohol drinking behavior and provide a unique pharmacologic target for reducing alcohol consumption.
Abstract: Excessive alcohol consumption is a major public health problem worldwide. Although drinking habits are known to be inherited, few genes have been identified that are robustly linked to alcohol drinking. We conducted a genome-wide association metaanalysis and replication study among >105,000 individuals of European ancestry and identified β-Klotho (KLB) as a locus associated with alcohol consumption (rs11940694; P = 9.2 × 10−12). β-Klotho is an obligate coreceptor for the hormone FGF21, which is secreted from the liver and implicated in macronutrient preference in humans. We show that brain-specific β-Klotho KO mice have an increased alcohol preference and that FGF21 inhibits alcohol drinking by acting on the brain. These data suggest that a liver–brain endocrine axis may play an important role in the regulation of alcohol drinking behavior and provide a unique pharmacologic target for reducing alcohol consumption.

205 citations


Journal ArticleDOI
Jessica N. Cooke Bailey1, Stephanie Loomis2, Jae H. Kang2, R. Rand Allingham3, Puya Gharahkhani4, Chiea Chuen Khor5, Chiea Chuen Khor6, Kathryn P. Burdon7, Kathryn P. Burdon8, Hugues Aschard2, Daniel I. Chasman2, Robert P. Igo1, Pirro G. Hysi9, Craig A. Glastonbury9, Allison E. Ashley-Koch3, Murray H. Brilliant10, Andrew A. Brown11, Donald L. Budenz12, Alfonso Buil11, Ching-Yu Cheng5, Ching-Yu Cheng13, Hyon K. Choi14, William G. Christen2, Gary C. Curhan2, Immaculata De Vivo2, John H. Fingert15, Paul J. Foster16, Paul J. Foster17, Charles S. Fuchs2, Douglas E. Gaasterland, Terry Gaasterland18, Alex W. Hewitt19, Frank B. Hu2, David J. Hunter2, Anthony P Khawaja20, Richard K. Lee21, Zheng Li6, Paul R. Lichter22, David A. Mackey23, David A. Mackey24, Peter McGuffin9, Paul Mitchell25, Sayoko E. Moroi22, Shamira A. Perera13, Shamira A. Perera5, Keating W. Pepper, Qibin Qi26, Tony Realini27, Julia E. Richards22, Paul M. Ridker2, Eric B. Rimm2, Robert Ritch28, Marylyn D. Ritchie29, Joel S. Schuman30, William K. Scott21, Kuldev Singh31, Arthur J. Sit32, Yeunjoo E. Song1, Rulla M. Tamimi2, Fotis Topouzis33, Ananth C. Viswanathan16, Shefali S. Verma29, Douglas Vollrath31, Jie Jin Wang25, Nicole Weisschuh34, Bernd Wissinger34, Gadi Wollstein30, Tien Yin Wong5, Tien Yin Wong13, Brian L. Yaspan35, Donald J. Zack36, Kang Zhang18, Robert N. Weinreb18, Margaret A. Pericak-Vance21, Kerrin S. Small9, Christopher J Hammond9, Tin Aung5, Tin Aung13, Yutao Liu37, Eranga N. Vithana5, Eranga N. Vithana13, Stuart MacGregor4, Jamie E Craig8, Peter Kraft2, Gareth R. Howell, Michael A. Hauser3, Louis R. Pasquale2, Jonathan L. Haines1, Janey L. Wiggs2 
TL;DR: New pathways underlying POAG susceptibility are identified and new targets for preventative therapies are suggested, including TXNRD2 and ATXN2 expression in retinal ganglion cells and the optic nerve head.
Abstract: Primary open-angle glaucoma (POAG) is a leading cause of blindness worldwide. To identify new susceptibility loci, we performed meta-analysis on genome-wide association study (GWAS) results from eight independent studies from the United States (3,853 cases and 33,480 controls) and investigated the most significantly associated SNPs in two Australian studies (1,252 cases and 2,592 controls), three European studies (875 cases and 4,107 controls) and a Singaporean Chinese study (1,037 cases and 2,543 controls). A meta-analysis of the top SNPs identified three new associated loci: rs35934224[T] in TXNRD2 (odds ratio (OR) = 0.78, P = 4.05 × 10(-11)) encoding a mitochondrial protein required for redox homeostasis; rs7137828[T] in ATXN2 (OR = 1.17, P = 8.73 × 10(-10)); and rs2745572[A] upstream of FOXC1 (OR = 1.17, P = 1.76 × 10(-10)). Using RT-PCR and immunohistochemistry, we show TXNRD2 and ATXN2 expression in retinal ganglion cells and the optic nerve head. These results identify new pathways underlying POAG susceptibility and suggest new targets for preventative therapies.


Journal ArticleDOI
TL;DR: In this article, the authors performed a genome-wide association study (GWAS) of circulating leptin levels from 32,161 individuals and identified five loci robustly associated with leptin levels in/near LEP, SLC32A1, GCKR, CCNL1 and FTO.
Abstract: Leptin is an adipocyte-secreted hormone, the circulating levels of which correlate closely with overall adiposity. Although rare mutations in the leptin (LEP) gene are well known to cause leptin deficiency and severe obesity, no common loci regulating circulating leptin levels have been uncovered. Therefore, we performed a genome-wide association study (GWAS) of circulating leptin levels from 32,161 individuals and followed up loci reaching P<10(-6) in 19,979 additional individuals. We identify five loci robustly associated (P<5 × 10(-8)) with leptin levels in/near LEP, SLC32A1, GCKR, CCNL1 and FTO. Although the association of the FTO obesity locus with leptin levels is abolished by adjustment for BMI, associations of the four other loci are independent of adiposity. The GCKR locus was found associated with multiple metabolic traits in previous GWAS and the CCNL1 locus with birth weight. Knockdown experiments in mouse adipose tissue explants show convincing evidence for adipogenin, a regulator of adipocyte differentiation, as the novel causal gene in the SLC32A1 locus influencing leptin levels. Our findings provide novel insights into the regulation of leptin production by adipose tissue and open new avenues for examining the influence of variation in leptin levels on adiposity and metabolic health.

Journal ArticleDOI
TL;DR: The ages of puberty, first sexual intercourse and first birth signify the onset of reproductive ability, behavior and success, respectively and Mendelian randomization analyses infer causal influences of earlier puberty timing on earlier firstSexual intercourse, earlier first birth and lower educational attainment.
Abstract: The ages of puberty, first sexual intercourse and first birth signify the onset of reproductive ability, behavior and success, respectively. In a genome-wide association study of 125,667 UK Biobank participants, we identify 38 loci associated (P < 5 × 10(-8)) with age at first sexual intercourse. These findings were taken forward in 241,910 men and women from Iceland and 20,187 women from the Women's Genome Health Study. Several of the identified loci also exhibit associations (P < 5 × 10(-8)) with other reproductive and behavioral traits, including age at first birth (variants in or near ESR1 and RBM6-SEMA3F), number of children (CADM2 and ESR1), irritable temperament (MSRA) and risk-taking propensity (CADM2). Mendelian randomization analyses infer causal influences of earlier puberty timing on earlier first sexual intercourse, earlier first birth and lower educational attainment. In turn, likely causal consequences of earlier first sexual intercourse include reproductive, educational, psychiatric and cardiometabolic outcomes.

Journal ArticleDOI
Ganesh Chauhan1, Corey R. Arnold2, Audrey Y. Chu, Myriam Fornage, Azadeh Reyahi, Joshua C. Bis, Aki S. Havulinna, Muralidharan Sargurupremraj, Albert V. Smith, Hieab H.H. Adams, Seung Hoan Choi, Sara L. Pulit, Stella Trompet, Melissa E. Garcia, Ani Manichaikul, Alexander Teumer, Stefan Gustafsson, Traci M. Bartz, Céline Bellenguez, Jean-Sébastien Vidal, Xueqiu Jian, Olafur Kjartansson, Kerri L. Wiggins, Claudia L. Satizabal, Flora Xue, Samuli Ripatti, Yongmei Liu, Joris Deelen, Marcel den Hoed, Steve Bevan, Jemma C. Hopewell, Rainer Malik, Susan R. Heckbert, Kenneth Rice, Nicholas L. Smith, Christopher R Levi, Pankaj Sharma, Cathie Sudlow, Ali Moussavi Nik, John W. Cole, Reinhold E. Schmidt, James F. Meschia, Vincent Thijs, Arne Lindgren2, Olle Melander, Raji P. Grewal, Ralph L. Sacco, Tatjana Rundek, Peter M. Rothwell, Donna K. Arnett, Christina Jern, Julie A. Johnson, Oscar R. Benavente, Sylvia Wassertheil-Smoller, Jin-Moo Lee, Quenna Wong, Hugo J. Aparicio, Stefan T. Engelter, Manja Kloss, Didier Leys, Alessandro Pezzini, Julie E. Buring, Paul M. Ridker, Claudine Berr, Jean-François Dartigues, Anders Hamsten, Patrik K. E. Magnusson, Matthew Traylor, Nancy L. Pedersen, Lars Lannfelt, Lars Lindgren, Cecilia M. Lindgren, Andrew P. Morris, Jordi Jimenez-Conde, Joan Montaner, Farid Radmanesh, Agnieszka Slowik, Daniel Woo, Albert Hofman, Peter J. Koudstaal, Marileen L.P. Portegies, André G. Uitterlinden, Anton J. M. de Craen, Ian Ford, J. Wouter Jukema, David J. Stott, Norrina B. Allen, Michèle M. Sale, Andrew D. Johnson, David A. Bennett, Philip L. De Jager, Charles C. White, Hans J. Grabe, Marcello Ricardo Paulista Markus, Ulf Schminke, Giorgio B. Boncoraglio, Robert Clarke, Yoichiro Kamatani, Jean Dallongeville, Oscar L. Lopez, Jerome I. Rotter, Mike A. Nalls, Rebecca F. Gottesman, Michael Griswold, David S. Knopman, B. Gwen Windham, Alexa S. Beiser, Hugh S. Markus, Erkki Vartiainen, Curtis R. French, Martin Dichgans, Tomi Pastinen, Mark Lathrop, Vilmundur Gudnason, Tobias Kurth, Bruce M. Psaty, Tamara B. Harris, Stephen S. Rich, Anita L. DeStefano, Carsten Oliver Schmidt, Bradford B. Worrall, Jonathan Rosand, Veikko Salomaa, Thomas H. Mosley, Erik Ingelsson, Cornelia M. van Duijn, Christophe Tzourio, Kathryn M. Rexrode, Ordan J. Lehmann, Lenore J. Launer, M. Arfan Ikram, Peter Carlsson, Daniel I. Chasman, Sarah J. Childs, William T. Longstreth, Sudha Seshadri, Stéphanie Debette1 
TL;DR: Empirical and experimental data suggest that FOXF2 mediates this association, potentially via differentiation defects of cerebral vascular mural cells, and further expression studies in appropriate human tissues are needed to fully understand the underlying mechanisms.
Abstract: Summary Background Genetic determinants of stroke, the leading neurological cause of death and disability, are poorly understood and have seldom been explored in the general population. Our aim was to identify additional loci for stroke by doing a meta-analysis of genome-wide association studies. Methods For the discovery sample, we did a genome-wide analysis of common genetic variants associated with incident stroke risk in 18 population-based cohorts comprising 84 961 participants, of whom 4348 had stroke. Stroke diagnosis was ascertained and validated by the study investigators. Mean age at stroke ranged from 45·8 years to 76·4 years, and data collection in the studies took place between 1948 and 2013. We did validation analyses for variants yielding a significant association (at p −6 ) with all-stroke, ischaemic stroke, cardioembolic ischaemic stroke, or non-cardioembolic ischaemic stroke in the largest available cross-sectional studies (70 804 participants, of whom 19 816 had stroke). Summary-level results of discovery and follow-up stages were combined using inverse-variance weighted fixed-effects meta-analysis, and in-silico lookups were done in stroke subtypes. For genome-wide significant findings (at p −8 ), we explored associations with additional cerebrovascular phenotypes and did functional experiments using conditional (inducible) deletion of the probable causal gene in mice. We also studied the expression of orthologs of this probable causal gene and its effects on cerebral vasculature in zebrafish mutants. Findings We replicated seven of eight known loci associated with risk for ischaemic stroke, and identified a novel locus at chromosome 6p25 (rs12204590, near FOXF2 ) associated with risk of all-stroke (odds ratio [OR] 1·08, 95% CI 1·05–1·12, p=1·48 × 10 −8 ; minor allele frequency 21%). The rs12204590 stroke risk allele was also associated with increased MRI-defined burden of white matter hyperintensity—a marker of cerebral small vessel disease—in stroke-free adults (n=21 079; p=0·0025). Consistently, young patients (aged 2–32 years) with segmental deletions of FOXF2 showed an extensive burden of white matter hyperintensity. Deletion of Foxf2 in adult mice resulted in cerebral infarction, reactive gliosis, and microhaemorrhage. The orthologs of FOXF2 in zebrafish (foxf2b and foxf2a ) are expressed in brain pericytes and mutant foxf2b −/− cerebral vessels show decreased smooth muscle cell and pericyte coverage. Interpretation We identified common variants near FOXF2 that are associated with increased stroke susceptibility. Epidemiological and experimental data suggest that FOXF2 mediates this association, potentially via differentiation defects of cerebral vascular mural cells. Further expression studies in appropriate human tissues, and further functional experiments with long follow-up periods are needed to fully understand the underlying mechanisms. Funding NIH, NINDS, NHMRC, CIHR, European national research institutions, Fondation Leducq.

Journal ArticleDOI
Carla A. Ibrahim-Verbaas1, Jan Bressler2, Stéphanie Debette, Maaike Schuur1, Albert V. Smith3, J. C. Bis4, Gary Davies5, Stella Trompet6, Jennifer A. Smith7, Christiane Wolf8, Lori B. Chibnik9, Yongmei Liu10, Veronique Vitart5, Mirna Kirin5, Katja Petrovic11, Ozren Polasek12, Lina Zgaga13, Chloe Fawns-Ritchie5, Peter Hoffmann14, Peter Hoffmann15, Juha Karjalainen16, Jari Lahti17, David J. Llewellyn18, Carsten Oliver Schmidt19, Karen A. Mather20, Vincent Chouraki21, Qi Sun9, Susan M. Resnick22, Lynda M. Rose9, Christopher Oldmeadow23, Marlene C. W. Stewart5, Blair H. Smith24, Vilmundur Gudnason3, Qiong Yang25, Saira Saeed Mirza1, Johan Wouter Jukema6, P. L. DeJager9, T.B. Harris22, D C Liewald5, Najaf Amin1, Laura H. Coker10, Oliver Stegle8, Oscar L. Lopez26, Reinhold Schmidt11, Alexander Teumer19, Ian Ford27, Nazanin Karbalai8, James T. Becker26, Maria K. Jonsdottir, Rhoda Au22, Rudolf S N Fehrmann16, Stefan Herms14, Stefan Herms15, Mike A. Nalls22, Wei Zhao7, Stephen T. Turner28, Kristine Yaffe29, Kurt Lohman10, J. C. van Swieten1, Sharon L.R. Kardia7, D. S. Knopman28, William M. Meeks30, Gerardo Heiss31, Elizabeth G. Holliday23, Peter W. Schofield23, Toshiko Tanaka22, David J. Stott27, Jing Wang25, Paul M. Ridker9, Alan J. Gow5, Alison Pattie5, John M. Starr5, Lynne J. Hocking32, Nicola J. Armstrong20, Stela McLachlan5, Joshua M. Shulman33, Luke C. Pilling18, G. Eiriksdottir, Rodney J. Scott23, Nicole A. Kochan20, Aarno Palotie17, Y. C. Hsieh34, Johan G. Eriksson, Alan D. Penman30, Rebecca F. Gottesman35, Ben A. Oostra1, Le Yu36, Anita L. DeStefano25, Alexa S. Beiser25, Melissa Garcia22, Jerome I. Rotter37, Jerome I. Rotter38, Markus M. Nöthen39, Albert Hofman1, P.E. Slagboom6, R.G.J. Westendorp, Brendan M. Buckley40, Philip A. Wolf22, André G. Uitterlinden1, Bruce M. Psaty41, Hans-Jörgen Grabe19, Stefania Bandinelli, Daniel I. Chasman9, Francine Grodstein9, Katri Räikkönen17, Jean-Charles Lambert21, David J. Porteous5, Jenna Price5, Perminder S. Sachdev20, Luigi Ferrucci22, John Attia23, Igor Rudan5, Caroline Hayward5, Alan F. Wright5, James F. Wilson5, Sven Cichon, Lude Franke16, Helena Schmidt11, Jun Ding10, A.J.M. de Craen6, Myriam Fornage2, David A. Bennett36, Ian J. Deary5, M. A. Ikram1, L. J. Launer22, Annette L. Fitzpatrick4, Sudha Seshadri22, C M van Duijn1, Thomas H. Mosley30 
TL;DR: This article conducted a GWAS of executive functioning and information processing speed in non-demented older adults from the CHARGE (Cohorts for Heart and Aging Research in Genomic Epidemiology) consortium.
Abstract: To identify common variants contributing to normal variation in two specific domains of cognitive functioning, we conducted a genome-wide association study (GWAS) of executive functioning and information processing speed in non-demented older adults from the CHARGE (Cohorts for Heart and Aging Research in Genomic Epidemiology) consortium. Neuropsychological testing was available for 5429-32,070 subjects of European ancestry aged 45 years or older, free of dementia and clinical stroke at the time of cognitive testing from 20 cohorts in the discovery phase. We analyzed performance on the Trail Making Test parts A and B, the Letter Digit Substitution Test (LDST), the Digit Symbol Substitution Task (DSST), semantic and phonemic fluency tests, and the Stroop Color and Word Test. Replication was sought in 1311-21860 subjects from 20 independent cohorts. A significant association was observed in the discovery cohorts for the single-nucleotide polymorphism (SNP) rs17518584 (discovery P-value=3.12 × 10(-8)) and in the joint discovery and replication meta-analysis (P-value=3.28 × 10(-9) after adjustment for age, gender and education) in an intron of the gene cell adhesion molecule 2 (CADM2) for performance on the LDST/DSST. Rs17518584 is located about 170 kb upstream of the transcription start site of the major transcript for the CADM2 gene, but is within an intron of a variant transcript that includes an alternative first exon. The variant is associated with expression of CADM2 in the cingulate cortex (P-value=4 × 10(-4)). The protein encoded by CADM2 is involved in glutamate signaling (P-value=7.22 × 10(-15)), gamma-aminobutyric acid (GABA) transport (P-value=1.36 × 10(-11)) and neuron cell-cell adhesion (P-value=1.48 × 10(-13)). Our findings suggest that genetic variation in the CADM2 gene is associated with individual differences in information processing speed.

Carla A. Ibrahim-Verbaas, Jan Bressler, Stéphanie Debette, Maaike Schuur, Albert V. Smith, J. C. Bis, G. Davies, S. Trompet, Jennifer A. Smith, Christiane Wolf, Lori B. Chibnik, Yongmei Liu, Veronique Vitart, Mirna Kirin, K. Petrovic, Ozren Polasek, Lina Zgaga, Chloe Fawns-Ritchie, Peter Hoffmann, Juha Karjalainen, Jari Lahti, David J. Llewellyn, Christian Schmidt, Karen A. Mather, Vincent Chouraki, Qi Sun, Susan M. Resnick, Ludger Rose, Christopher Oldmeadow, Marlene C. W. Stewart, Blair H. Smith, Vilmundur Gudnason, Qiong Yang, Saira Saeed Mirza, Joop Jukema, Philip L. DeJager, T.B. Harris, D C Liewald, Najaf Amin, Laura H. Coker, Oliver Stegle, Oscar L. Lopez, R. Schmidt, A. Teumer, Ian Ford, Nazanin Karbalai, James T. Becker, Maria K. Jonsdottir, Rhoda Au, Rudolf S N Fehrmann, Stefan Herms, Michael A. Nalls, Wei Zhao, Stephen T. Turner, Kristine Yaffe, Kurt Lohman, J. C. van Swieten, Sharon L.R. Kardia, D. S. Knopman, William M. Meeks, Gerardo Heiss, Elizabeth G. Holliday, Peter W. Schofield, Toshiko Tanaka, David J. Stott, Jing Wang, Paul M. Ridker, Alan J. Gow, Alison Pattie, John M. Starr, Lynne J. Hocking, Nicola J. Armstrong, Stela McLachlan, Joshua M. Shulman, Luke C. Pilling, G. Eiriksdottir, Rodney J. Scott, Nicole A. Kochan, Aarno Palotie, Y-C Hsieh, Johan G. Eriksson, Alan D. Penman, Rebecca F. Gottesman, B.A. Oostra, Le Yu, Anita L. DeStefano, Alexa S. Beiser, Melissa Garcia, Jerome I. Rotter, Markus M. Noethen, Albert Hofman, P.E. Slagboom, R.G.J. Westendorp, Brendan M. Buckley, Philip A. Wolf, André G. Uitterlinden, Bruce M. Psaty, Hans-Jörgen Grabe, Stefania Bandinelli, Daniel I. Chasman, Francine Grodstein, K. Roikkonen, J-C Lambert, David J. Porteous, Jenna Price, Perminder S. Sachdev, L Ferrucci, John Attia, Igor Rudan, Caroline Hayward, Antony Wright, James F. Wilson, Sven Cichon, Lude Franke, H. Schmidt, Jun Ding, A.J.M. de Craen, Myriam Fornage, David A. Bennett, Ian J. Deary, M. A. Ikram, L. J. Launer, Annette L. Fitzpatrick, Sudha Seshadri, C M van Duijn, Thomas H. Mosley 
17 Mar 2016
TL;DR: A genome-wide association study of executive functioning and information processing speed in non-demented older adults from the CHARGE consortium suggests that genetic variation in the CADM2 gene is associated with individual differences in informationprocessing speed.
Abstract: To identify common variants contributing to normal variation in two specific domains of cognitive functioning, we conducted a genome-wide association study (GWAS) of executive functioning and information processing speed in non-demented older adults from the CHARGE (Cohorts for Heart and Aging Research in Genomic Epidemiology) consortium. Neuropsychological testing was available for 5429–32 070 subjects of European ancestry aged 45 years or older, free of dementia and clinical stroke at the time of cognitive testing from 20 cohorts in the discovery phase. We analyzed performance on the Trail Making Test parts A and B, the Letter Digit Substitution Test (LDST), the Digit Symbol Substitution Task (DSST), semantic and phonemic fluency tests, and the Stroop Color and Word Test. Replication was sought in 1311-21860 subjects from 20 independent cohorts. A significant association was observed in the discovery cohorts for the single-nucleotide polymorphism (SNP) rs17518584 (discovery P-value=3.12 × 10−8) and in the joint discovery and replication meta-analysis (P-value=3.28 × 10−9 after adjustment for age, gender and education) in an intron of the gene cell adhesion molecule 2 (CADM2) for performance on the LDST/DSST. Rs17518584 is located about 170 kb upstream of the transcription start site of the major transcript for the CADM2 gene, but is within an intron of a variant transcript that includes an alternative first exon. The variant is associated with expression of CADM2 in the cingulate cortex (P-value=4 × 10−4). The protein encoded by CADM2 is involved in glutamate signaling (P-value=7.22 × 10−15), gamma-aminobutyric acid (GABA) transport (P-value=1.36 × 10−11) and neuron cell-cell adhesion (P-value=1.48 × 10−13). Our findings suggest that genetic variation in the CADM2 gene is associated with individual differences in information processing speed.

Padhraig Gormley, Verneri Anttila, Bendik S. Winsvold, Priit Palta, Tõnu Esko, Tune H. Pers, K.-H. Farh, E. Cuenca-Leon, Mikko Muona, N.A. Furlotte, Tobias Kurth, Andres Ingason, Gearoid M. McMahon, Lannie Ligthart, Gisela M. Terwindt, Mikko Kallela, T. Freilinger, Caroline Ran, G. Gordon, AH Stam, Stacy Steinberg, Guntram Borck, Markku Koiranen, Lydia Quaye, Hieab H.H. Adams, Terho Lehtimäki, Antti-Pekka Sarin, Juho Wedenoja, David A. Hinds, Julie E. Buring, Markus Schürks, Paul M. Ridker, M.G. Hrafnsdottir, Hreinn Stefansson, S.M. Ring, Jouke-Jan Hottenga, Brenda W.J.H. Penninx, Markus Färkkilä, Ville Artto, Mari A. Kaunisto, Salli Vepsäläinen, Rainer Malik, A. C. Heath, P. A. F. Madden, Glynn Martin, Grant W. Montgomery, Eija Hämäläinen, Hailiang Huang, Andrea Byrnes, Lude Franke, Jie Huang, Evie Stergiakouli, Phil Lee, Cynthia Sandor, Caleb Webber, Zameel Cader, Bertram Müller-Myhsok, Stefan Schreiber, Thomas Meitinger, Johan G. Eriksson, V. Salomaa, Kauko Heikkilä, Elizabeth Loehrer, André G. Uitterlinden, Albert Hofman, C M van Duijn, Lynn Cherkas, Linda M. Pedersen, Audun Stubhaug, Christopher Sivert Nielsen, Minna Männikkö, Evelin Mihailov, Lili Milani, H. Gobel, A.-L. Esserlind, Annette Christensen, Thomas Hansen, Thomas Werge, Jaakko Kaprio, Arpo Aromaa, Olli T. Raitakari, Ikram, Tim D. Spector, Marjo-Riitta Järvelin, Andres Metspalu, Christian Kubisch, David P. Strachan, Ferrari, Andrea Carmine Belin, Martin Dichgans, Maija Wessman, A.M.J.M. van den Maagdenberg, John-Anker Zwart, Dorret I. Boomsma, George Davey Smith, K. Stefansson, Nicholas Eriksson, Mark J. Daly, Benjamin M. Neale, Jes Olesen, Daniel I. Chasman, Dale R. Nyholt, Aarno Palotie, Lyn R. Griffiths, Huiying Zhao 
01 Jun 2016
TL;DR: For example, the authors identified 45 independent single nucleotide polymorphisms (SNPs) significantly associated with migraine risk that map to 38 distinct genomic loci, including 28 loci not previously reported and the first locus identified on chromosome X.
Abstract: Migraine is a debilitating neurological disorder affecting around 1 in 7 people worldwide, but its molecular mechanisms remain poorly understood. Some debate exists over whether migraine is a disease of vascular dysfunction, or a result of neuronal dysfunction with secondary vascular changes. Genome-wide association (GWA) studies have thus far identified 13 independent loci associated with migraine. To identify new susceptibility loci, we performed the largest genetic study of migraine to date, comprising 59,674 cases and 316,078 controls from 22 GWA studies. We identified 45 independent single nucleotide polymorphisms (SNPs) significantly associated with migraine risk ( P -8 ) that map to 38 distinct genomic loci, including 28 loci not previously reported and the first locus identified on chromosome X. Furthermore, a subset analysis for migraine without aura (MO) identified seven of the same loci as from the full sample, whereas no loci reached genome-wide significance in the migraine with aura (MA) subset. In subsequent computational analyzes, the identified loci showed enrichment for genes expressed in vascular and smooth muscle tissues, consistent with a predominant theory of migraine that highlights vascular etiologies.

Journal ArticleDOI
Paul S. de Vries, Daniel I. Chasman1, Daniel I. Chasman2, Maria Sabater-Lleal, Ming-Huei Chen3, Jennifer E. Huffman4, Maristella Steri, Weihong Tang, Alexander Teumer, Riccardo E. Marioni5, Vera Grossmann, Jouke J. Hottenga6, Stella Trompet, Martina Müller-Nurasyid7, Jing Hua Zhao, Jennifer A. Brody, Marcus E. Kleber8, Xiuqing Guo9, Jie Jin Wang10, Paul L. Auer11, John Attia, Lisa R. Yanek12, Tarunveer S. Ahluwalia13, Jari Lahti, Cristina Venturini14, Toshiko Tanaka4, Lawrence F. Bielak15, Peter K. Joshi, Ares Rocanin-Arjo16, Ivana Kolcic, Pau Navarro, Lynda M. Rose1, Christopher Oldmeadow, Helene Riess, Johanna Mazur, Saonli Basu17, Anuj Goel18, Qiong Yang3, Mohsen Ghanbari19, Gonneke Willemsen6, Ann Rumley, Edoardo Fiorillo, Anton J. M. de Craen, Anne Grotevendt, Robert A. Scott, Kent D. Taylor, Graciela E. Delgado8, Jie Yao, Annette Kifley10, Charles Kooperberg20, Rehan Qayyum12, Lorna M. Lopez21, Lorna M. Lopez22, Tina L. Berentzen23, Katri Räikkönen, Massimo Mangino14, Stefania Bandinelli, Patricia A. Peyser15, Sarah H. Wild, David-Alexandre Trégouët16, Alan F. Wright, Jonathan Marten, Tatijana Zemunik, Alanna C. Morrison24, Bengt Sennblad25, Geoffrey H. Tofler26, Moniek P.M. de Maat, Eco J. C. de Geus6, Gordon D.O. Lowe, Magdalena Zoledziewska, Naveed Sattar27, Harald Binder, Uwe Völker, Melanie Waldenberger, Kay-Tee Khaw28, Barbara McKnight, Jie Huang29, Nancy S. Jenny, Elizabeth G. Holliday, Lihong Qi30, Mark G. Mcevoy31, Diane M. Becker12, John M. Starr, Antti-Pekka Sarin4, Antti-Pekka Sarin32, Pirro G. Hysi14, Dena G. Hernandez4, Min A. Jhun15, Harry Campbell, Anders Hamsten, Fernando Rivadeneira33, Wendy L. McArdle34, P. Eline Slagboom35, Tanja Zeller36, Wolfgang Koenig37, Bruce M. Psaty38, Talin Haritunians39, Jingmin Liu20, Aarno Palotie32, André G. Uitterlinden33, David J. Stott27, Albert Hofman, Oscar H. Franco, Ozren Polasek40, Igor Rudan, Pierre-Emmanuel Morange41, James F. Wilson, Sharon L.R. Kardia15, Luigi Ferrucci4, Tim D. Spector14, Johan G. Eriksson, Torben Hansen42, Ian J. Deary43, Lewis C. Becker12, Rodney J. Scott31, Paul Mitchell10, Winfried März44, Nicholas J. Wareham, Annette Peters, Andreas Greinacher45, Philipp S. Wild46, J. Wouter Jukema, Dorret I. Boomsma6, Caroline Hayward, Francesco Cucca, Russell P. Tracy47, Hugh Watkins18, Alexander P. Reiner20, Aaron R. Folsom, Paul M. Ridker2, Paul M. Ridker1, Christopher J. O'Donnell2, Nicholas L. Smith48, David P. Strachan49, Abbas Dehghan 
Brigham and Women's Hospital1, Harvard University2, Boston University3, National Institutes of Health4, University of Queensland5, VU University Amsterdam6, Ludwig Maximilian University of Munich7, Heidelberg University8, Los Angeles Biomedical Research Institute9, University of Sydney10, University of Wisconsin–Milwaukee11, Johns Hopkins University School of Medicine12, University of Copenhagen13, King's College London14, University of Michigan15, Pierre-and-Marie-Curie University16, University of Minnesota17, Wellcome Trust Centre for Human Genetics18, Mashhad University of Medical Sciences19, Fred Hutchinson Cancer Research Center20, University College Dublin21, Royal College of Surgeons in Ireland22, Frederiksberg Hospital23, University of Texas Health Science Center at Houston24, Science for Life Laboratory25, Royal North Shore Hospital26, University of Glasgow27, University of Cambridge28, Wellcome Trust Sanger Institute29, University of California, Davis30, University of Newcastle31, University of Helsinki32, Erasmus University Rotterdam33, University of Bristol34, Leiden University Medical Center35, University of Hamburg36, Technische Universität München37, Group Health Research Institute38, Cedars-Sinai Medical Center39, University of Split40, Aix-Marseille University41, Novo Nordisk Foundation42, University of Edinburgh43, Medical University of Graz44, Greifswald University Hospital45, University of Mainz46, University of Vermont47, University of Washington48, St George's, University of London49
TL;DR: A genome-wide association analysis of 34 studies imputed to the 1000 Genomes Project reference panel and including ∼120 000 participants of European ancestry identified 41 genome- wide significant fibrinogen loci; of which, 18 were newly identified.
Abstract: Genome-wide association studies have previously identified 23 genetic loci associated with circulating fibrinogen concentration. These studies used HapMap imputation and did not examine the X-chromosome. 1000 Genomes imputation provides better coverage of uncommon variants, and includes indels. We conducted a genome-wide association analysis of 34 studies imputed to the 1000 Genomes Project reference panel and including ∼120 000 participants of European ancestry (95 806 participants with data on the X-chromosome). Approximately 10.7 million single-nucleotide polymorphisms and 1.2 million indels were examined. We identified 41 genome-wide significant fibrinogen loci; of which, 18 were newly identified. There were no genome-wide significant signals on the X-chromosome. The lead variants of five significant loci were indels. We further identified six additional independent signals, including three rare variants, at two previously characterized loci: FGB and IRF1. Together the 41 loci explain 3% of the variance in plasma fibrinogen concentration.

Journal ArticleDOI
TL;DR: The results of the analysis show that inflammatory responses initiate from within the cross-talk of the three identified endophenotypic modules, and are also enriched with differentially expressed genes linked to cardiovascular disease (risk).
Abstract: Historically, human diseases have been differentiated and categorized based on the organ system in which they primarily manifest. Recently, an alternative view is emerging that emphasizes that different diseases often have common underlying mechanisms and shared intermediate pathophenotypes, or endo(pheno)types. Within this framework, a specific disease's expression is a consequence of the interplay between the relevant endophenotypes and their local, organ-based environment. Important examples of such endophenotypes are inflammation, fibrosis, and thrombosis and their essential roles in many developing diseases. In this study, we construct endophenotype network models and explore their relation to different diseases in general and to cardiovascular diseases in particular. We identify the local neighborhoods (module) within the interconnected map of molecular components, i.e., the subnetworks of the human interactome that represent the inflammasome, thrombosome, and fibrosome. We find that these neighborhoods are highly overlapping and significantly enriched with disease-associated genes. In particular they are also enriched with differentially expressed genes linked to cardiovascular disease (risk). Finally, using proteomic data, we explore how macrophage activation contributes to our understanding of inflammatory processes and responses. The results of our analysis show that inflammatory responses initiate from within the cross-talk of the three identified endophenotypic modules.

Journal ArticleDOI
07 Mar 2016-PLOS ONE
TL;DR: The association of the 9p21 locus both with increased risk of first myocardial infarction and longer survival after MI highlights the importance of study design in investigating genetic determinants of complex disorders.
Abstract: Background Data are limited on genome-wide association studies (GWAS) for incident coronary heart disease (CHD). Moreover, it is not known whether genetic variants identified to date also associate with risk of CHD in a prospective setting. Methods We performed a two-stage GWAS analysis of incident myocardial infarction (MI) and CHD in a total of 64,297 individuals (including 3898 MI cases, 5465 CHD cases). SNPs that passed an arbitrary threshold of 5x10(-6) in Stage I were taken to Stage II for further discovery. Furthermore, in an analysis of prognosis, we studied whether known SNPs from former GWAS were associated with total mortality in individuals who experienced MI during follow-up. Results In Stage I 15 loci passed the threshold of 5x10(-6); 8 loci for MI and 8 loci for CHD, for which one locus overlapped and none were reported in previous GWAS meta-analyses. We took 60 SNPs representing these 15 loci to Stage II of discovery. Four SNPs near QKI showed nominally significant association with MI (p-value<8.8x10(-3)) and three exceeded the genome-wide significance threshold when Stage I and Stage II results were combined (top SNP rs6941513: p = 6.2x10(-9)). Despite excellent power, the 9p21 locus SNP (rs1333049) was only modestly associated with MI (HR = 1.09, p-value = 0.02) and marginally with CHD (HR = 1.06, p-value = 0.08). Among an inception cohort of those who experienced MI during follow-up, the risk allele of rs1333049 was associated with a decreased risk of subsequent mortality (HR = 0.90, p-value = 3.2x10(-3)). Conclusions QKI represents a novel locus that may serve as a predictor of incident CHD in prospective studies. The association of the 9p21 locus both with increased risk of first myocardial infarction and longer survival after MI highlights the importance of study design in investigating genetic determinants of complex disorders.

Journal ArticleDOI
TL;DR: In this paper, the authors examined whether genetic variants affect body shape as a composite phenotype that is represented by a combination of anthropometric traits, and identified six novel loci: LEMD2 and CD47 for AvPC1, RPS6KA5/C14orf159 and GANAB for AVPC3, and ARL15 and ANP32 for Avpc4.
Abstract: Large consortia have revealed hundreds of genetic loci associated with anthropometric traits, one trait at a time. We examined whether genetic variants affect body shape as a composite phenotype that is represented by a combination of anthropometric traits. We developed an approach that calculates averaged PCs (AvPCs) representing body shape derived from six anthropometric traits (body mass index, height, weight, waist and hip circumference, waist-to-hip ratio). The first four AvPCs explain >99% of the variability, are heritable, and associate with cardiometabolic outcomes. We performed genome-wide association analyses for each body shape composite phenotype across 65 studies and meta-analysed summary statistics. We identify six novel loci: LEMD2 and CD47 for AvPC1, RPS6KA5/C14orf159 and GANAB for AvPC3, and ARL15 and ANP32 for AvPC4. Our findings highlight the value of using multiple traits to define complex phenotypes for discovery, which are not captured by single-trait analyses, and may shed light onto new pathways.

Journal ArticleDOI
TL;DR: A large-scale, meta-analysis of GWASs of individuals of European ancestry with available prior genotype data identified 4 loci in genes that have putative functions in cholesterol metabolism and transport, and sulfonylation of bile acids or hydroxysteroids.

Journal ArticleDOI
TL;DR: The observed polygenic overlap between CAD and cardiometabolic risk factors indicates a pathogenic relation that warrants further investigation.
Abstract: Rationale: Coronary Artery Disease (CAD) is a critical determinant of morbidity and mortality. Previous studies have identified several cardiovascular disease (CVD) risk factors, which may partly arise from a shared genetic basis with CAD, and thus be useful for discovery of CAD genes. Objective: We aimed to improve discovery of CAD genes, and inform the etiologic relationship between CAD and several CVD risk factors using a shared polygenic signal-informed statistical framework. Methods and Results: Using genome-wide association studies (GWAS) summary statistics and shared polygenic pleiotropy-informed conditional and conjunctional false discovery rate (FDR) methodology, we systematically investigated genetic overlap between CAD and 8 traits related to CVD risk factors: low density lipoprotein (LDL) cholesterol, high density lipoprotein (HDL) cholesterol, triglycerides (TG), type 2 diabetes (T2D), C-reactive protein (CRP), body mass index (BMI), systolic blood pressure (SBP) and type 1 diabetes (T1D). We found significant enrichment of single nucleotide polymorphisms (SNPs) associated with CAD as a function of their association with LDL, HDL, TG, T2D, CRP, BMI, SBP and T1D. Applying the conditional FDR method to the enriched phenotypes, we identified 67 novel loci associated with CAD (overall conditional FDR < 0.01). Further, we identified 53 loci with significant effects in both CAD and at least one of LDL, HDL, TG, T2D, CRP, SBP and T1D. Conclusions: The observed polygenic overlap between CAD and cardio-metabolic risk factors indicates an etiological relation that warrants further investigation. The new gene loci identified implicate novel genetic mechanisms related to CAD.

Journal ArticleDOI
Nathan O. Stitziel, Kathleen Stirrups, Nicholas G. D. Masca, Jeanette Erdmann, Paola G. Ferrario, Inke R. Koenig, Peter Weeke, Tom R. Webb, Paul L. Auer, Ursula M. Schick, Yingchang Lu, He Zhang, Marie-Pierre Dubé, Anuj Goel, Martin Farrall, Gina M. Peloso, Hong-Hee Won, Ron Do, Erik P A Van Iperen, Stavroula Kanoni, Jochen Kruppa, Anubha Mahajan, Robert A. Scott, Christina Willenborg, Peter S. Braund, Julian C. van Capelleveen, Alex S. F. Doney, Louise A. Donnelly, Rosanna Asselta, Piera Angelica Merlini, Stefano Duga, Nicola Marziliano, Josh C. Denny, Christian M. Shaffer, Nour Eddine El-Mokhtari, Andre Franke, Omri Gottesman, Stefanie Heilmann, Christian Hengstenberg, Per Hoffmann, Oddgeir L. Holmen, Kristian Hveem, Jan-Håkan Jansson, Karl-Heinz Joeckel, Thorsten Kessler, Jennifer Kriebel, Karl L. Laugwitz, Eirini Marouli, Nicola Martinelli, Mark I. McCarthy, Natalie R. van Zuydam, Christa Meisinger, Tõnu Esko, Evelin Mihailov, Stefan A. Escher, Maris Alver, Susanne Moebus, Andrew D. Morris, Martina Müller-Nurasyid, Majid Nikpay, Oliviero Olivieri, Louis-Philippe Lemieux Perreault, Alaa AlQarawi, Neil Robertson, Karen O. Akinsanya, Dermot F. Reilly, Thomas F. Vogt, Wu Yin, Folkert W. Asselbergs, Charles Kooperberg, Rebecca D. Jackson, Eli A. Stahl, Konstantin Strauch, Tibor V. Varga, Melanie Waldenberger, Lingyao Zeng, Aldi T. Kraja, Chunyu Liu, Georg Ehret, Christopher Newton-Cheh, Daniel I. Chasman, Rajiv Chowdhury, Marco M Ferrario, Ian Ford, J. Wouter Jukema, Frank Kee, Kari Kuulasmaa, Børge G. Nordestgaard, Markus Perola, Danish Saleheen, Naveed Sattar, Praveen Surendran, David-Alexandre Trégouët, Robin Young, Joanna M. M. Howson, Adam S. Butterworth, John Danesh, Diego Ardissino, Erwin P. Bottinger, Raimund Erbel, Paul W. Franks, Domenico Girelli, Alistair S. Hall, G. Kees Hovingh, Adnan Kastrati, Wolfgang Lieb, Thomas Meitinger, William E. Kraus, Svati H. Shah, Ruth McPherson, Marju Orho-Melander, Olle Melander, Andres Metspalu, Colin N. A. Palmer, Annette Peters, Daniel J. Rader, Muredach P. Reilly, Ruth J. F. Loos, Alex P. Reiner, Dan M. Roden, Jean-Claude Tardif, John Thompson, Nicholas J. Wareham, Hugh Watkins, Cristen J. Willer, Sekar Kathiresan, Panos Deloukas, Nilesh J. Samani, Heribert Schunkert 

Journal ArticleDOI
TL;DR: This work integrated migraine GWAS data with high-resolution spatial gene expression data of normal adult brains from the Allen Human Brain Atlas to identify specific brain regions and molecular pathways that are possibly involved in migraine pathophysiology and used the high-confidence genes from the GWAS as a basis to construct local migraine-related co-expression gene networks.
Abstract: Migraine is a common disabling neurovascular brain disorder typically characterised by attacks of severe headache and associated with autonomic and neurological symptoms. Migraine is caused by an interplay of genetic and environmental factors. Genome-wide association studies (GWAS) have identified over a dozen genetic loci associated with migraine. Here, we integrated migraine GWAS data with high-resolution spatial gene expression data of normal adult brains from the Allen Human Brain Atlas to identify specific brain regions and molecular pathways that are possibly involved in migraine pathophysiology. To this end, we used two complementary methods. In GWAS data from 23,285 migraine cases and 95,425 controls, we first studied modules of co-expressed genes that were calculated based on human brain expression data for enrichment of genes that showed association with migraine. Enrichment of a migraine GWAS signal was found for five modules that suggest involvement in migraine pathophysiology of: (i) neurotransmission, protein catabolism and mitochondria in the cortex; (ii) transcription regulation in the cortex and cerebellum; and (iii) oligodendrocytes and mitochondria in subcortical areas. Second, we used the high-confidence genes from the migraine GWAS as a basis to construct local migraine-related co-expression gene networks. Signatures of all brain regions and pathways that were prominent in the first method also surfaced in the second method, thus providing support that these brain regions and pathways are indeed involved in migraine pathophysiology.

Journal ArticleDOI
TL;DR: In this paper, it is unclear whether patients diagnosed according to International Classification of Headache Disorders criteria for migraine with aura (MA) and migraine without aura (MO) experience d...
Abstract: IntroductionIt is unclear whether patients diagnosed according to International Classification of Headache Disorders criteria for migraine with aura (MA) and migraine without aura (MO) experience d...

01 Oct 2016
TL;DR: A large genome-wide association study of both sexes including 251,151 individuals for AFB and 343,072 individuals for NEB finds 12 independent loci that are significantly associated with AFB and/or NEB in a SNP-based genome- wide association study and 4 additional loci associated in a gene-based effort.
Abstract: The genetic architecture of human reproductive behavior—age at first birth (AFB) and number of children ever born (NEB)—has a strong relationship with fitness, human development, infertility and risk of neuropsychiatric disorders. However, very few genetic loci have been identified, and the underlying mechanisms of AFB and NEB are poorly understood. We report a large genome-wide association study of both sexes including 251,151 individuals for AFB and 343,072 individuals for NEB. We identified 12 independent loci that are significantly associated with AFB and/or NEB in a SNP-based genome-wide association study and 4 additional loci associated in a gene-based effort. These loci harbor genes that are likely to have a role, either directly or by affecting non-local gene expression, in human reproduction and infertility, thereby increasing understanding of these complex traits.

Journal ArticleDOI
TL;DR: This integrative study is the first to associate rs76481776 with POAG via elevated miR-182 expression using miRNA sequencing and ddPCR, and it is unclear whether this association was primarily in the HTG subset.
Abstract: Author(s): Liu, Yutao; Bailey, Jessica Cooke; Helwa, Inas; Dismuke, W Michael; Cai, Jingwen; Drewry, Michelle; Brilliant, Murray H; Budenz, Donald L; Christen, William G; Chasman, Daniel I; Fingert, John H; Gaasterland, Douglas; Gaasterland, Terry; Gordon, Mae O; Igo, Robert P; Kang, Jae H; Kass, Michael A; Kraft, Peter; Lee, Richard K; Lichter, Paul; Moroi, Sayoko E; Realini, Anthony; Richards, Julia E; Ritch, Robert; Schuman, Joel S; Scott, William K; Singh, Kuldev; Sit, Arthur J; Song, Yeunjoo E; Vollrath, Douglas; Weinreb, Robert; Medeiros, Felipe; Wollstein, Gadi; Zack, Donald J; Zhang, Kang; Pericak-Vance, Margaret A; Gonzalez, Pedro; Stamer, W Daniel; Kuchtey, John; Kuchtey, Rachel W; Allingham, R Rand; Hauser, Michael A; Pasquale, Louis R; Haines, Jonathan L; Wiggs, Janey L | Abstract: PurposeNoncoding microRNAs (miRNAs) have been implicated in the pathogenesis of glaucoma. We aimed to identify common variants in miRNA coding genes (MIR) associated with primary open-angle glaucoma (POAG).MethodsUsing the NEIGHBORHOOD data set (3853 cases/33,480 controls with European ancestry), we first assessed the relation between 85 variants in 76 MIR genes and overall POAG. Subtype-specific analyses were performed in high-tension glaucoma (HTG) and normal-tension glaucoma subsets. Second, we examined the expression of miR-182, which was associated with POAG, in postmortem human ocular tissues (ciliary body, cornea, retina, and trabecular meshwork [TM]), using miRNA sequencing (miRNA-Seq) and droplet digital PCR (ddPCR). Third, miR-182 expression was also examined in human aqueous humor (AH) by using miRNA-Seq. Fourth, exosomes secreted from primary human TM cells were examined for miR-182 expression by using miRNA-Seq. Fifth, using ddPCR we compared miR-182 expression in AH between five HTG cases and five controls.ResultsOnly rs76481776 in MIR182 gene was associated with POAG after adjustment for multiple comparisons (odds ratio [OR] = 1.23, 95% confidence interval [CI]: 1.11-1.42, P = 0.0002). Subtype analysis indicated that the association was primarily in the HTG subset (OR = 1.26, 95% CI: 1.08-1.47, P = 0.004). The risk allele T has been associated with elevated miR-182 expression in vitro. Data from ddPCR and miRNA-Seq confirmed miR-182 expression in all examined ocular tissues and TM-derived exosomes. Interestingly, miR-182 expression in AH was 2-fold higher in HTG patients than nonglaucoma controls (P = 0.03) without controlling for medication treatment.ConclusionsOur integrative study is the first to associate rs76481776 with POAG via elevated miR-182 expression.

Journal ArticleDOI
TL;DR: It is indicated that common, low frequency and rare CHRNA5 coding variants are independently associated with nicotine dependence risk and likely influence the risk for smoking-related diseases such as lung cancer.
Abstract: The common nonsynonymous variant rs16969968 in the α5 nicotinic receptor subunit gene (CHRNA5) is the strongest genetic risk factor for nicotine dependence in European Americans and contributes to risk in African Americans. To comprehensively examine whether other CHRNA5 coding variation influences nicotine dependence risk, we performed targeted sequencing on 1582 nicotine-dependent cases (Fagerstrom Test for Nicotine Dependence score⩾4) and 1238 non-dependent controls, with independent replication of common and low frequency variants using 12 studies with exome chip data. Nicotine dependence was examined using logistic regression with individual common variants (minor allele frequency (MAF)⩾0.05), aggregate low frequency variants (0.05>MAF⩾0.005) and aggregate rare variants (MAF<0.005). Meta-analysis of primary results was performed with replication studies containing 12 174 heavy and 11 290 light smokers. Next-generation sequencing with 180 × coverage identified 24 nonsynonymous variants and 2 frameshift deletions in CHRNA5, including 9 novel variants in the 2820 subjects. Meta-analysis confirmed the risk effect of the only common variant (rs16969968, European ancestry: odds ratio (OR)=1.3, P=3.5 × 10(-11); African ancestry: OR=1.3, P=0.01) and demonstrated that three low frequency variants contributed an independent risk (aggregate term, European ancestry: OR=1.3, P=0.005; African ancestry: OR=1.4, P=0.0006). The remaining 22 rare coding variants were associated with increased risk of nicotine dependence in the European American primary sample (OR=12.9, P=0.01) and in the same risk direction in African Americans (OR=1.5, P=0.37). Our results indicate that common, low frequency and rare CHRNA5 coding variants are independently associated with nicotine dependence risk. These newly identified variants likely influence the risk for smoking-related diseases such as lung cancer.