151
|
Brown AA, Viñuela A, Delaneau O, Spector TD, Small KS, Dermitzakis ET. Predicting causal variants affecting expression by using whole-genome sequencing and RNA-seq from multiple human tissues. Nat Genet 2017; 49:1747-1751. [PMID: 29058714 DOI: 10.1038/ng.3979] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 09/27/2017] [Indexed: 12/16/2022]
Abstract
Genetic association mapping produces statistical links between phenotypes and genomic regions, but identifying causal variants remains difficult. Whole-genome sequencing (WGS) can help by providing complete knowledge of all genetic variants, but it is financially prohibitive for well-powered GWAS studies. We performed mapping of expression quantitative trait loci (eQTLs) with WGS and RNA-seq, and found that lead eQTL variants called with WGS were more likely to be causal. Through simulations, we derived properties of causal variants and used them to develop a method for identifying likely causal SNPs. We estimated that 25-70% of causal variants were located in open-chromatin regions, depending on the tissue and experiment. Finally, we identified a set of high-confidence causal variants and showed that these were more enriched in GWAS associations than other eQTLs. Of those, we found 65 associations with GWAS traits and provide examples in which genes implicated by expression are functionally validated as being relevant for complex traits.
Collapse
Affiliation(s)
- Andrew Anand Brown
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland.,Swiss Institute of Bioinformatics, Geneva, Switzerland.,NORMENT, KG Jebsen Centre for Psychosis Research, Oslo University Hospital, Oslo, Norway
| | - Ana Viñuela
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland.,Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Olivier Delaneau
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland.,Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland.,Swiss Institute of Bioinformatics, Geneva, Switzerland
| |
Collapse
|
152
|
CCDC88B is required for pathogenesis of inflammatory bowel disease. Nat Commun 2017; 8:932. [PMID: 29030607 PMCID: PMC5640600 DOI: 10.1038/s41467-017-01381-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 09/13/2017] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel disease (IBD) involves interaction between host genetic factors and environmental triggers. CCDC88B maps within one IBD risk locus on human chromosome 11q13. Here we show that CCDC88B protein increases in the colon during intestinal injury, concomitant with an influx of CCDC88B+lymphoid and myeloid cells. Loss of Ccdc88b protects against DSS-induced colitis, with fewer pathological lesions and reduced intestinal inflammation in Ccdc88b-deficient mice. In a T cell transfer model of colitis, Ccdc88b mutant CD4+ T cells do not induce colitis in immunocompromised hosts. Expression of human CCDC88B RNA and protein is higher in IBD patient colons than in control colon tissue. In human CD14+ myeloid cells, CCDC88B is regulated by cis-acting variants. In a cohort of patients with Crohn's disease, CCDC88B expression correlates positively with disease risk. These findings suggest that CCDC88B has a critical function in colon inflammation and the pathogenesis of IBD.Hook-related protein family member CCDC88b is encoded by a locus that has been associated with inflammatory bowel disease. Here the authors show that Ccdc88b inactivation in T cells prevents colitis in a transfer model, and detect high colonic levels of CCDC88b in patients with Crohn disease or ulcerative colitis, identifying that expression correlates with disease risk.
Collapse
|
153
|
Carvalho TL, Lima RE, Góes GHB, Pereira LA, Fernandes MSDS, Moura PMMF, Vasconcelos LRS, Correia CC. Cognitive Dysfunction and Single Nucleotide Polymorphisms in Hepatitis C Virus-Infected Persons: A Systematic Review. Viral Immunol 2017; 30:703-707. [PMID: 29016246 DOI: 10.1089/vim.2017.0084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The aim of this study was to realize a systematic review to identify data reported in the literature involving people infected by hepatitis C virus (HCV) with cognitive dysfunctions and single nucleotide polymorphisms (SNPs). The research was realized in six databases and the selection of studies was performed in two stages. Initially, we searched indexed articles from the following electronic databases: SciELO, MEDLINE, PubMed, HighWire, LILACS, and ScienceDirect. Then the articles were completely read and those that did not meet the eligibility criteria were excluded. Therefore, 5,669 articles were obtained and, of these, 25 were selected. Finally, one article involving people with HCV and cognitive impairment was included in the review. The frequency of the APOE-ɛ4 allele in people with HCV and mild liver disease was significantly lower in those with work memory impairment (p = 0.003) and attention (p = 0.008). This situation differs from other studies that showed an association between ɛ4 allele high frequency and cognitive decline. Thus, studies with larger samples involving people with HCV, cognitive alterations, and SNPs are necessary, in view of the lack of this theme in the literature and the divergences in the findings.
Collapse
Affiliation(s)
- Tatiana Lins Carvalho
- 1 Hospital Universitário Oswaldo Cruz, Universidade de Pernambuco (UPE) , Recife, PE, Brazil
| | - Raul Emídio Lima
- 2 Instituto de Ciências Biológicas, Universidade de Pernambuco (UPE) , Recife, PE, Brazil
| | | | - Lívio Amaro Pereira
- 3 Medical Sciences College, Universidade de Pernambuco (UPE) , Recife, PE, Brazil
| | | | | | | | - Carolina Cunha Correia
- 6 Hospital Universitário Oswaldo Cruz; Medical Sciences College, Universidade de Pernambuco (UPE) , Recife, PE, Brazil
| |
Collapse
|
154
|
Odhams CA, Cunninghame Graham DS, Vyse TJ. Profiling RNA-Seq at multiple resolutions markedly increases the number of causal eQTLs in autoimmune disease. PLoS Genet 2017; 13:e1007071. [PMID: 29059182 PMCID: PMC5695635 DOI: 10.1371/journal.pgen.1007071] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 11/02/2017] [Accepted: 10/11/2017] [Indexed: 01/12/2023] Open
Abstract
Genome-wide association studies have identified hundreds of risk loci for autoimmune disease, yet only a minority (~25%) share genetic effects with changes to gene expression (eQTLs) in immune cells. RNA-Seq based quantification at whole-gene resolution, where abundance is estimated by culminating expression of all transcripts or exons of the same gene, is likely to account for this observed lack of colocalisation as subtle isoform switches and expression variation in independent exons can be concealed. We performed integrative cis-eQTL analysis using association statistics from twenty autoimmune diseases (560 independent loci) and RNA-Seq data from 373 individuals of the Geuvadis cohort profiled at gene-, isoform-, exon-, junction-, and intron-level resolution in lymphoblastoid cell lines. After stringently testing for a shared causal variant using both the Joint Likelihood Mapping and Regulatory Trait Concordance frameworks, we found that gene-level quantification significantly underestimated the number of causal cis-eQTLs. Only 5.0-5.3% of loci were found to share a causal cis-eQTL at gene-level compared to 12.9-18.4% at exon-level and 9.6-10.5% at junction-level. More than a fifth of autoimmune loci shared an underlying causal variant in a single cell type by combining all five quantification types; a marked increase over current estimates of steady-state causal cis-eQTLs. Causal cis-eQTLs detected at different quantification types localised to discrete epigenetic annotations. We applied a linear mixed-effects model to distinguish cis-eQTLs modulating all expression elements of a gene from those where the signal is only evident in a subset of elements. Exon-level analysis detected disease-associated cis-eQTLs that subtly altered transcription globally across the target gene. We dissected in detail the genetic associations of systemic lupus erythematosus and functionally annotated the candidate genes. Many of the known and novel genes were concealed at gene-level (e.g. IKZF2, TYK2, LYST). Our findings are provided as a web resource.
Collapse
Affiliation(s)
- Christopher A. Odhams
- Department of Medical & Molecular Genetics, King’s College London, London, United Kingdom
| | - Deborah S. Cunninghame Graham
- Department of Medical & Molecular Genetics, King’s College London, London, United Kingdom
- Academic Department of Rheumatology, Division of Immunology, Infection and Inflammatory Disease, King’s College London, London, United Kingdom
| | - Timothy J. Vyse
- Department of Medical & Molecular Genetics, King’s College London, London, United Kingdom
- Academic Department of Rheumatology, Division of Immunology, Infection and Inflammatory Disease, King’s College London, London, United Kingdom
| |
Collapse
|
155
|
Dissecting genetic architecture of startle response in Drosophila melanogaster using multi-omics information. Sci Rep 2017; 7:12367. [PMID: 28959013 PMCID: PMC5620086 DOI: 10.1038/s41598-017-11676-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 08/24/2017] [Indexed: 01/01/2023] Open
Abstract
Startle behavior is important for survival, and abnormal startle responses are related to several neurological diseases. Drosophila melanogaster provides a powerful system to investigate the genetic underpinnings of variation in startle behavior. Since mechanically induced, startle responses and environmental conditions can be readily quantified and precisely controlled. The 156 wild-derived fully sequenced lines of the Drosophila Genetic Reference Panel (DGRP) were used to identify SNPs and transcripts associated with variation in startle behavior. The results validated highly significant effects of 33 quantitative trait SNPs (QTSs) and 81 quantitative trait transcripts (QTTs) directly associated with phenotypic variation of startle response. We also detected QTT variation controlled by 20 QTSs (tQTSs) and 73 transcripts (tQTTs). Association mapping based on genomic and transcriptomic data enabled us to construct a complex genetic network that underlies variation in startle behavior. Based on principles of evolutionary conservation, human orthologous genes could be superimposed on this network. This study provided both genetic and biological insights into the variation of startle response behavior of Drosophila melanogaster, and highlighted the importance of genetic network to understand the genetic architecture of complex traits.
Collapse
|
156
|
Gaspar L, Howald C, Popadin K, Maier B, Mauvoisin D, Moriggi E, Gutierrez-Arcelus M, Falconnet E, Borel C, Kunz D, Kramer A, Gachon F, Dermitzakis ET, Antonarakis SE, Brown SA. The genomic landscape of human cellular circadian variation points to a novel role for the signalosome. eLife 2017; 6:e24994. [PMID: 28869038 PMCID: PMC5601996 DOI: 10.7554/elife.24994] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 09/01/2017] [Indexed: 11/18/2022] Open
Abstract
The importance of natural gene expression variation for human behavior is undisputed, but its impact on circadian physiology remains mostly unexplored. Using umbilical cord fibroblasts, we have determined by genome-wide association how common genetic variation impacts upon cellular circadian function. Gene set enrichment points to differences in protein catabolism as one major source of clock variation in humans. The two most significant alleles regulated expression of COPS7B, a subunit of the COP9 signalosome. We further show that the signalosome complex is imported into the nucleus in timed fashion to stabilize the essential circadian protein BMAL1, a novel mechanism to oppose its proteasome-mediated degradation. Thus, circadian clock properties depend in part upon a genetically-encoded competition between stabilizing and destabilizing forces, and genetic alterations in these mechanisms provide one explanation for human chronotype.
Collapse
Affiliation(s)
- Ludmila Gaspar
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| | - Cedric Howald
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in GenevaUniversity of GenevaGenevaSwitzerland
| | - Konstantin Popadin
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenevaSwitzerland
| | - Bert Maier
- Charité–UniversitätsmedizinLaboratory of ChronobiologyBerlinGermany
| | - Daniel Mauvoisin
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
| | - Ermanno Moriggi
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| | - Maria Gutierrez-Arcelus
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in GenevaUniversity of GenevaGenevaSwitzerland
| | - Emilie Falconnet
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in GenevaUniversity of GenevaGenevaSwitzerland
| | - Christelle Borel
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in GenevaUniversity of GenevaGenevaSwitzerland
| | - Dieter Kunz
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Working Group Sleep Research & Clinical ChronobiologyBerlinGermany
| | - Achim Kramer
- Charité–UniversitätsmedizinLaboratory of ChronobiologyBerlinGermany
| | - Frederic Gachon
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in GenevaUniversity of GenevaGenevaSwitzerland
| | - Stylianos E Antonarakis
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in GenevaUniversity of GenevaGenevaSwitzerland
| | - Steven A Brown
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
157
|
|
158
|
Schieffer KM, Choi CS, Emrich S, Harris L, Deiling S, Karamchandani DM, Salzberg A, Kawasawa YI, Yochum GS, Koltun WA. RNA-seq implicates deregulation of the immune system in the pathogenesis of diverticulitis. Am J Physiol Gastrointest Liver Physiol 2017; 313:G277-G284. [PMID: 28619727 PMCID: PMC6146301 DOI: 10.1152/ajpgi.00136.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 01/31/2023]
Abstract
Individuals with diverticula or outpouchings of the colonic mucosa and submucosa through the colonic wall have diverticulosis, which is usually asymptomatic. In 10-25% of individuals, the diverticula become inflamed, resulting in diverticulitis. Very little is known about the pathophysiology or gene regulatory pathways involved in the development of diverticulitis. To identify these pathways, we deep sequenced RNAs isolated from full-thickness sections of sigmoid colon from diverticulitis patients and control individuals. Specifically for diverticulitis cases, we analyzed tissue adjacent to areas affected by chronic disease. Since the tissue was collected during elective sigmoid resection, the disease was in a quiescent state. A comparison of differentially expressed genes found that gene ontology (GO) pathways associated with the immune response were upregulated in diverticulitis patients compared with nondiverticulosis controls. Next, weighted gene coexpression network analysis was performed to identify the interaction among coexpressed genes. This analysis revealed RASAL3, SASH3, PTPRC, and INPP5D as hub genes within the brown module eigengene, which highly correlated (r = 0.67, P = 0.0004) with diverticulitis. Additionally, we identified elevated expression of downstream interacting genes. In summary, transcripts associated with the immune response were upregulated in adjacent tissue from the sigmoid colons of chronic, recurrent diverticulitis patients. Further elucidating the genetic or epigenetic mechanisms associated with these alterations can help identify those at risk for chronic disease and may assist in clinical decision management.NEW & NOTEWORTHY By using an unbiased approach to analyze transcripts expressed in unaffected colonic tissues adjacent to those affected by chronic diverticulitis, our study implicates that a defect in the immune response may be involved in the development of the disease. This finding expands on the current data that suggest the pathophysiology of diverticulitis is mediated by dietary, age, and obesity-related factors. Further characterizing the immunologic differences in diverticulitis may better inform clinical decision-making.
Collapse
Affiliation(s)
- Kathleen M Schieffer
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Christine S Choi
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Scott Emrich
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Leonard Harris
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Sue Deiling
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Dipti M Karamchandani
- Division of Anatomic Pathology, Department of Pathology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Anna Salzberg
- Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Yuka I Kawasawa
- Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and
| | - Gregory S Yochum
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Walter A Koltun
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania;
| |
Collapse
|
159
|
Constraints on eQTL Fine Mapping in the Presence of Multisite Local Regulation of Gene Expression. G3-GENES GENOMES GENETICS 2017; 7:2533-2544. [PMID: 28600440 PMCID: PMC5555460 DOI: 10.1534/g3.117.043752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Expression quantitative trait locus (eQTL) detection has emerged as an important tool for unraveling of the relationship between genetic risk factors and disease or clinical phenotypes. Most studies use single marker linear regression to discover primary signals, followed by sequential conditional modeling to detect secondary genetic variants affecting gene expression. However, this approach assumes that functional variants are sparsely distributed and that close linkage between them has little impact on estimation of their precise location and the magnitude of effects. We describe a series of simulation studies designed to evaluate the impact of linkage disequilibrium (LD) on the fine mapping of causal variants with typical eQTL effect sizes. In the presence of multisite regulation, even though between 80 and 90% of modeled eSNPs associate with normally distributed traits, up to 10% of all secondary signals could be statistical artifacts, and at least 5% but up to one-quarter of credible intervals of SNPs within r2 > 0.8 of the peak may not even include a causal site. The Bayesian methods eCAVIAR and DAP (Deterministic Approximation of Posteriors) provide only modest improvement in resolution. Given the strong empirical evidence that gene expression is commonly regulated by more than one variant, we conclude that the fine mapping of causal variants needs to be adjusted for multisite influences, as conditional estimates can be highly biased by interference among linked sites, but ultimately experimental verification of individual effects is needed. Presumably similar conclusions apply not just to eQTL mapping, but to multisite influences on fine mapping of most types of quantitative trait.
Collapse
|
160
|
Elkon R, Agami R. Characterization of noncoding regulatory DNA in the human genome. Nat Biotechnol 2017; 35:732-746. [DOI: 10.1038/nbt.3863] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 03/31/2017] [Indexed: 12/22/2022]
|
161
|
Huang H, Fang M, Jostins L, Umićević Mirkov M, Boucher G, Anderson CA, Andersen V, Cleynen I, Cortes A, Crins F, D'Amato M, Deffontaine V, Dmitrieva J, Docampo E, Elansary M, Farh KKH, Franke A, Gori AS, Goyette P, Halfvarson J, Haritunians T, Knight J, Lawrance IC, Lees CW, Louis E, Mariman R, Meuwissen T, Mni M, Momozawa Y, Parkes M, Spain SL, Théâtre E, Trynka G, Satsangi J, van Sommeren S, Vermeire S, Xavier RJ, Weersma RK, Duerr RH, Mathew CG, Rioux JD, McGovern DPB, Cho JH, Georges M, Daly MJ, Barrett JC. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 2017; 547:173-178. [PMID: 28658209 PMCID: PMC5511510 DOI: 10.1038/nature22969] [Citation(s) in RCA: 408] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 05/07/2017] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel diseases are chronic gastrointestinal inflammatory disorders that affect millions of people worldwide. Genome-wide association studies have identified 200 inflammatory bowel disease-associated loci, but few have been conclusively resolved to specific functional variants. Here we report fine-mapping of 94 inflammatory bowel disease loci using high-density genotyping in 67,852 individuals. We pinpoint 18 associations to a single causal variant with greater than 95% certainty, and an additional 27 associations to a single variant with greater than 50% certainty. These 45 variants are significantly enriched for protein-coding changes (n = 13), direct disruption of transcription-factor binding sites (n = 3), and tissue-specific epigenetic marks (n = 10), with the last category showing enrichment in specific immune cells among associations stronger in Crohn's disease and in gut mucosa among associations stronger in ulcerative colitis. The results of this study suggest that high-resolution fine-mapping in large samples can convert many discoveries from genome-wide association studies into statistically convincing causal variants, providing a powerful substrate for experimental elucidation of disease mechanisms.
Collapse
Affiliation(s)
- Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA
| | - Ming Fang
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Luke Jostins
- Wellcome Trust Centre for Human Genetics, University of Oxford, Headington OX3 7BN, UK.,Christ Church, University of Oxford, St Aldates OX1 1DP, UK
| | - Maša Umićević Mirkov
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Gabrielle Boucher
- Research Center, Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada
| | - Carl A Anderson
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Vibeke Andersen
- Focused research unit for Molecular Diagnostic and Clinical Research (MOK), IRS-Center Sonderjylland, Hospital of Southern Jutland, 6200 Åbenrå, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | | | - Adrian Cortes
- Wellcome Trust Centre for Human Genetics, University of Oxford, Headington OX3 7BN, UK.,Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - François Crins
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Mauro D'Amato
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden.,Department of Gastrointestinal and Liver Diseases, BioDonostia Health Research Institute, 20014 San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Valérie Deffontaine
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Julia Dmitrieva
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Elisa Docampo
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Mahmoud Elansary
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Kyle Kai-How Farh
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA.,Illumina, San Diego, California 92122, USA
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany
| | - Ann-Stephan Gori
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Philippe Goyette
- Research Center, Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, SE-70182 Örebro, Sweden
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Jo Knight
- Data Science Institute and Lancaster Medical School, Lancaster University, Lancaster LA1 4YG, UK
| | - Ian C Lawrance
- Centre for Inflammatory Bowel Diseases, Saint John of God Hospital, Subiaco, Western Australia 6008, Australia.,Harry Perkins Institute for Medical Research, School of Medicine and Pharmacology, University of Western Australia, Murdoch, Western Australia 6150, Australia
| | - Charlie W Lees
- Gastrointestinal Unit, Western General Hospital University of Edinburgh, Edinburgh, UK
| | - Edouard Louis
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Division of Gastroenterology, Centre Hospitalier Universitaire (CHU) de Liège, 4000 Liège, Belgium
| | - Rob Mariman
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Theo Meuwissen
- Institute of Livestock and Aquacultural Sciences, Norwegian University of Life Sciences, 1430 Ås, Norway
| | - Myriam Mni
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Yukihide Momozawa
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium.,Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama, Kanagawa 230-0045, Japan
| | - Miles Parkes
- Inflammatory Bowel Disease Research Group, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Sarah L Spain
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK.,Open Targets, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Emilie Théâtre
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Gosia Trynka
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Jack Satsangi
- Gastrointestinal Unit, Western General Hospital University of Edinburgh, Edinburgh, UK
| | - Suzanne van Sommeren
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, 9700RB Groningen, The Netherlands
| | - Severine Vermeire
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium.,Division of Gastroenterology, University Hospital Gasthuisberg, 3000 Leuven, Belgium
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA.,Gastroenterology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, 9700RB Groningen, The Netherlands
| | - Richard H Duerr
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania 15261, USA
| | - Christopher G Mathew
- Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, UK.,Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - John D Rioux
- Research Center, Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada.,Faculté de Médecine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Dermot P B McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Judy H Cho
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Michel Georges
- Unit of Medical Genomics, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-R) Research Center and WELBIO, University of Liège, 4000 Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA
| | - Jeffrey C Barrett
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| |
Collapse
|
162
|
Do C, Shearer A, Suzuki M, Terry MB, Gelernter J, Greally JM, Tycko B. Genetic-epigenetic interactions in cis: a major focus in the post-GWAS era. Genome Biol 2017. [PMID: 28629478 PMCID: PMC5477265 DOI: 10.1186/s13059-017-1250-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Studies on genetic-epigenetic interactions, including the mapping of methylation quantitative trait loci (mQTLs) and haplotype-dependent allele-specific DNA methylation (hap-ASM), have become a major focus in the post-genome-wide-association-study (GWAS) era. Such maps can nominate regulatory sequence variants that underlie GWAS signals for common diseases, ranging from neuropsychiatric disorders to cancers. Conversely, mQTLs need to be filtered out when searching for non-genetic effects in epigenome-wide association studies (EWAS). Sequence variants in CCCTC-binding factor (CTCF) and transcription factor binding sites have been mechanistically linked to mQTLs and hap-ASM. Identifying these sites can point to disease-associated transcriptional pathways, with implications for targeted treatment and prevention.
Collapse
Affiliation(s)
- Catherine Do
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Alyssa Shearer
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Masako Suzuki
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University Mailman School of Public Health, and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Joel Gelernter
- Departments of Psychiatry, Genetics, and Neurobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - John M Greally
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Benjamin Tycko
- Institute for Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Taub Institute for Research on Alzheimer's disease and the Aging Brain, New York, NY, 10032, USA. .,Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
163
|
Hauberg ME, Zhang W, Giambartolomei C, Franzén O, Morris DL, Vyse TJ, Ruusalepp A, Sklar P, Schadt EE, Björkegren JL, Roussos P, Fromer M, Sieberts SK, Johnson JS, Ruderfer DM, Shah HR, Klei LL, Dang KK, Perumal TM, Logsdon BA, Mahajan MC, Mangravite LM, Essioux L, Toyoshiba H, Gur RE, Hahn CG, Lewis DA, Haroutunian V, Peters MA, Lipska BK, Buxbaum JD, Hirai K, Domenici E, Devlin B. Large-Scale Identification of Common Trait and Disease Variants Affecting Gene Expression. Am J Hum Genet 2017; 100:885-894. [PMID: 28552197 DOI: 10.1016/j.ajhg.2017.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/26/2017] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified a multitude of genetic loci involved with traits and diseases. However, it is often unclear which genes are affected in such loci and whether the associated genetic variants lead to increased or decreased gene function. To mitigate this, we integrated associations of common genetic variants in 57 GWASs with 24 studies of expression quantitative trait loci (eQTLs) from a broad range of tissues by using a Mendelian randomization approach. We discovered a total of 3,484 instances of gene-trait-associated changes in expression at a false-discovery rate < 0.05. These genes were often not closest to the genetic variant and were primarily identified in eQTLs derived from pathophysiologically relevant tissues. For instance, genes with expression changes associated with lipid traits were mostly identified in the liver, and those associated with cardiovascular disease were identified in arterial tissue. The affected genes additionally point to biological processes implicated in the interrogated traits, such as the interleukin-27 pathway in rheumatoid arthritis. Further, comparing trait-associated gene expression changes across traits suggests that pleiotropy is a widespread phenomenon and points to specific instances of both agonistic and antagonistic pleiotropy. For instance, expression of SNX19 and ABCB9 is positively correlated with both the risk of schizophrenia and educational attainment. To facilitate interpretation, we provide this lexicon of how common trait-associated genetic variants alter gene expression in various tissues as the online database GWAS2Genes.
Collapse
|
164
|
Markunas CA, Johnson EO, Hancock DB. Comprehensive evaluation of disease- and trait-specific enrichment for eight functional elements among GWAS-identified variants. Hum Genet 2017; 136:911-919. [PMID: 28567521 DOI: 10.1007/s00439-017-1815-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/22/2017] [Indexed: 01/17/2023]
Abstract
Genome-wide association study (GWAS)-identified variants are enriched for functional elements. However, we have limited knowledge of how functional enrichment may differ by disease/trait and tissue type. We tested a broad set of eight functional elements for enrichment among GWAS-identified SNPs (p < 5×10-8) from the NHGRI-EBI Catalog across seven disease/trait categories: cancer, cardiovascular disease, diabetes, autoimmune disease, psychiatric disease, neurological disease, and anthropometric traits. SNPs were annotated using HaploReg for the eight functional elements across any tissue: DNase sites, expression quantitative trait loci (eQTL), sequence conservation, enhancers, promoters, missense variants, sequence motifs, and protein binding sites. In addition, tissue-specific annotations were considered for brain vs. blood. Disease/trait SNPs were compared to a control set of 4809 SNPs matched to the GWAS SNPs (N = 1639) on allele frequency, gene density, distance to nearest gene, and linkage disequilibrium at ~3:1 ratio. Enrichment analyses were conducted using logistic regression, with Bonferroni correction. Overall, a significant enrichment was observed for all functional elements, except sequence motifs. Missense SNPs showed the strongest magnitude of enrichment. eQTLs were the only functional element significantly enriched across all diseases/traits. Magnitudes of enrichment were generally similar across diseases/traits, where enrichment was statistically significant. Blood vs. brain tissue effects on enrichment were dependent on disease/trait and functional element (e.g., cardiovascular disease: eQTLs P TissueDifference = 1.28 × 10-6 vs. enhancers P TissueDifference = 0.94). Identifying disease/trait-relevant functional elements and tissue types could provide new insight into the underlying biology, by guiding a priori GWAS analyses (e.g., brain enhancer elements for psychiatric disease) or facilitating post hoc interpretation.
Collapse
Affiliation(s)
- Christina A Markunas
- Behavioral Health and Criminal Justice Division, RTI International, Research Triangle Park, NC, USA.
| | - Eric O Johnson
- Behavioral Health and Criminal Justice Division, RTI International, Research Triangle Park, NC, USA.,Fellow Program, RTI International, Research Triangle Park, NC, USA
| | - Dana B Hancock
- Behavioral Health and Criminal Justice Division, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
165
|
Polygenic burdens on cell-specific pathways underlie the risk of rheumatoid arthritis. Nat Genet 2017; 49:1120-1125. [PMID: 28553958 DOI: 10.1038/ng.3885] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 05/03/2017] [Indexed: 12/15/2022]
Abstract
Recent evidence suggests that a substantial portion of complex disease risk alleles modify gene expression in a cell-specific manner. To identify candidate causal genes and biological pathways of immune-related complex diseases, we conducted expression quantitative trait loci (eQTL) analysis on five subsets of immune cells (CD4+ T cells, CD8+ T cells, B cells, natural killer (NK) cells and monocytes) and unfractionated peripheral blood from 105 healthy Japanese volunteers. We developed a three-step analytical pipeline comprising (i) prediction of individual gene expression using our eQTL database and public epigenomic data, (ii) gene-level association analysis and (iii) prediction of cell-specific pathway activity by integrating the direction of eQTL effects. By applying this pipeline to rheumatoid arthritis data sets, we identified candidate causal genes and a cytokine pathway (upregulation of tumor necrosis factor (TNF) in CD4+ T cells). Our approach is an efficient way to characterize the polygenic contributions and potential biological mechanisms of complex diseases.
Collapse
|
166
|
Delaneau O, Ongen H, Brown AA, Fort A, Panousis NI, Dermitzakis ET. A complete tool set for molecular QTL discovery and analysis. Nat Commun 2017; 8:15452. [PMID: 28516912 PMCID: PMC5454369 DOI: 10.1038/ncomms15452] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 03/30/2017] [Indexed: 12/26/2022] Open
Abstract
Population scale studies combining genetic information with molecular phenotypes (for example, gene expression) have become a standard to dissect the effects of genetic variants onto organismal phenotypes. These kinds of data sets require powerful, fast and versatile methods able to discover molecular Quantitative Trait Loci (molQTL). Here we propose such a solution, QTLtools, a modular framework that contains multiple new and well-established methods to prepare the data, to discover proximal and distal molQTLs and, finally, to integrate them with GWAS variants and functional annotations of the genome. We demonstrate its utility by performing a complete expression QTL study in a few easy-to-perform steps. QTLtools is open source and available at https://qtltools.github.io/qtltools/. Analysis of molecular quantitative trait loci (molQTL) can help interpret genome-wide association studies and requires efficient approaches to correct for multiple testing. This study describes a bioinformatics toolkit called QTLtool that can handle large data sets and quickly perform multiple types of molQTL analyses.
Collapse
Affiliation(s)
- Olivier Delaneau
- Department of Genetic Medicine and Development, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Swiss Institute of Bioinformatics, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Institute of Genetics and Genomics in Geneva, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland
| | - Halit Ongen
- Department of Genetic Medicine and Development, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Swiss Institute of Bioinformatics, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Institute of Genetics and Genomics in Geneva, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland
| | - Andrew A Brown
- Department of Genetic Medicine and Development, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Swiss Institute of Bioinformatics, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Institute of Genetics and Genomics in Geneva, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland
| | - Alexandre Fort
- Department of Genetic Medicine and Development, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland
| | - Nikolaos I Panousis
- Department of Genetic Medicine and Development, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Swiss Institute of Bioinformatics, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Institute of Genetics and Genomics in Geneva, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Swiss Institute of Bioinformatics, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland.,Institute of Genetics and Genomics in Geneva, University of Geneva, 1 Michel Servet, Geneva CH1211, Switzerland
| |
Collapse
|
167
|
Odhams CA, Cortini A, Chen L, Roberts AL, Viñuela A, Buil A, Small KS, Dermitzakis ET, Morris DL, Vyse TJ, Cunninghame Graham DS. Mapping eQTLs with RNA-seq reveals novel susceptibility genes, non-coding RNAs and alternative-splicing events in systemic lupus erythematosus. Hum Mol Genet 2017; 26:1003-1017. [PMID: 28062664 PMCID: PMC5409091 DOI: 10.1093/hmg/ddw417] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/05/2016] [Indexed: 12/19/2022] Open
Abstract
Studies attempting to functionally interpret complex-disease susceptibility loci by GWAS and eQTL integration have predominantly employed microarrays to quantify gene-expression. RNA-Seq has the potential to discover a more comprehensive set of eQTLs and illuminate the underlying molecular consequence. We examine the functional outcome of 39 variants associated with Systemic Lupus Erythematosus (SLE) through the integration of GWAS and eQTL data from the TwinsUK microarray and RNA-Seq cohort in lymphoblastoid cell lines. We use conditional analysis and a Bayesian colocalisation method to provide evidence of a shared causal-variant, then compare the ability of each quantification type to detect disease relevant eQTLs and eGenes. We discovered the greatest frequency of candidate-causal eQTLs using exon-level RNA-Seq, and identified novel SLE susceptibility genes (e.g. NADSYN1 and TCF7) that were concealed using microarrays, including four non-coding RNAs. Many of these eQTLs were found to influence the expression of several genes, supporting the notion that risk haplotypes may harbour multiple functional effects. Novel SLE associated splicing events were identified in the T-reg restricted transcription factor, IKZF2, and other candidate genes (e.g. WDFY4) through asQTL mapping using the Geuvadis cohort. We have significantly increased our understanding of the genetic control of gene-expression in SLE by maximising the leverage of RNA-Seq and performing integrative GWAS-eQTL analysis against gene, exon, and splice-junction quantifications. We conclude that to better understand the true functional consequence of regulatory variants, quantification by RNA-Seq should be performed at the exon-level as a minimum, and run in parallel with gene and splice-junction level quantification.
Collapse
Affiliation(s)
| | - Andrea Cortini
- Department of Medical & Molecular Genetics, King's College London, London, UK
| | - Lingyan Chen
- Department of Medical & Molecular Genetics, King's College London, London, UK
| | - Amy L Roberts
- Department of Medical & Molecular Genetics, King's College London, London, UK
| | - Ana Viñuela
- Department of Twin Research, King's College London, London, UK
| | | | - Kerrin S Small
- Department of Twin Research, King's College London, London, UK
| | | | - David L Morris
- Department of Medical & Molecular Genetics, King's College London, London, UK
| | - Timothy J Vyse
- Department of Medical & Molecular Genetics, King's College London, London, UK.,Division of Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| | | |
Collapse
|
168
|
Jansen R, Hottenga JJ, Nivard MG, Abdellaoui A, Laport B, de Geus EJ, Wright FA, Penninx BWJH, Boomsma DI. Conditional eQTL analysis reveals allelic heterogeneity of gene expression. Hum Mol Genet 2017; 26:1444-1451. [PMID: 28165122 DOI: 10.1093/hmg/ddx043] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/27/2017] [Indexed: 11/14/2022] Open
Abstract
In recent years, multiple eQTL (expression quantitative trait loci) catalogs have become available that can help understand the functionality of complex trait-related single nucleotide polymorphisms (SNPs). In eQTL catalogs, gene expression is often strongly associated with multiple SNPs, which may reflect either one or multiple independent associations. Conditional eQTL analysis allows a distinction between dependent and independent eQTLs. We performed conditional eQTL analysis in 4,896 peripheral blood microarray gene expression samples. Our analysis showed that 35% of genes with a cis eQTL have at least two independent cis eQTLs; for several genes up to 13 independent cis eQTLs were identified. Also, 12% (671) of the independent cis eQTLs identified in conditional analyses were not significant in unconditional analyses. The number of GWAS catalog SNPs identified as eQTL in the conditional analyses increases with 24% as compared to unconditional analyses. We provide an online conditional cis eQTL mapping catalog for whole blood (https://eqtl.onderzoek.io/), which can be used to lookup eQTLs more accurately than in standard unconditional whole blood eQTL databases.
Collapse
Affiliation(s)
- Rick Jansen
- Department of Psychiatry, Vrije Universiteit Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health, Amsterdam, The Netherlands
| | - Michel G Nivard
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health, Amsterdam, The Netherlands
| | - Abdel Abdellaoui
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health, Amsterdam, The Netherlands
| | - Bram Laport
- Department of Psychiatry, Vrije Universiteit Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Eco J de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health, Amsterdam, The Netherlands
| | - Fred A Wright
- Departments of Statistics and Biological Sciences, Bioinformatics Research Center, North Carolina State University, NC, USA
| | - Brenda W J H Penninx
- Department of Psychiatry, Vrije Universiteit Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health, Amsterdam, The Netherlands
| |
Collapse
|
169
|
Ju JH, Shenoy SA, Crystal RG, Mezey JG. An independent component analysis confounding factor correction framework for identifying broad impact expression quantitative trait loci. PLoS Comput Biol 2017; 13:e1005537. [PMID: 28505156 PMCID: PMC5448815 DOI: 10.1371/journal.pcbi.1005537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 05/30/2017] [Accepted: 04/28/2017] [Indexed: 11/19/2022] Open
Abstract
Genome-wide expression Quantitative Trait Loci (eQTL) studies in humans have provided numerous insights into the genetics of both gene expression and complex diseases. While the majority of eQTL identified in genome-wide analyses impact a single gene, eQTL that impact many genes are particularly valuable for network modeling and disease analysis. To enable the identification of such broad impact eQTL, we introduce CONFETI: Confounding Factor Estimation Through Independent component analysis. CONFETI is designed to address two conflicting issues when searching for broad impact eQTL: the need to account for non-genetic confounding factors that can lower the power of the analysis or produce broad impact eQTL false positives, and the tendency of methods that account for confounding factors to model broad impact eQTL as non-genetic variation. The key advance of the CONFETI framework is the use of Independent Component Analysis (ICA) to identify variation likely caused by broad impact eQTL when constructing the sample covariance matrix used for the random effect in a mixed model. We show that CONFETI has better performance than other mixed model confounding factor methods when considering broad impact eQTL recovery from synthetic data. We also used the CONFETI framework and these same confounding factor methods to identify eQTL that replicate between matched twin pair datasets in the Multiple Tissue Human Expression Resource (MuTHER), the Depression Genes Networks study (DGN), the Netherlands Study of Depression and Anxiety (NESDA), and multiple tissue types in the Genotype-Tissue Expression (GTEx) consortium. These analyses identified both cis-eQTL and trans-eQTL impacting individual genes, and CONFETI had better or comparable performance to other mixed model confounding factor analysis methods when identifying such eQTL. In these analyses, we were able to identify and replicate a few broad impact eQTL although the overall number was small even when applying CONFETI. In light of these results, we discuss the broad impact eQTL that have been previously reported from the analysis of human data and suggest that considerable caution should be exercised when making biological inferences based on these reported eQTL.
Collapse
Affiliation(s)
- Jin Hyun Ju
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States of America
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Sushila A. Shenoy
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Jason G. Mezey
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States of America
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, United States of America
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY, United States of America
- * E-mail:
| |
Collapse
|
170
|
Hormozdiari F, Zhu A, Kichaev G, Ju CJT, Segrè AV, Joo JWJ, Won H, Sankararaman S, Pasaniuc B, Shifman S, Eskin E. Widespread Allelic Heterogeneity in Complex Traits. Am J Hum Genet 2017; 100:789-802. [PMID: 28475861 PMCID: PMC5420356 DOI: 10.1016/j.ajhg.2017.04.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/07/2017] [Indexed: 12/24/2022] Open
Abstract
Recent successes in genome-wide association studies (GWASs) make it possible to address important questions about the genetic architecture of complex traits, such as allele frequency and effect size. One lesser-known aspect of complex traits is the extent of allelic heterogeneity (AH) arising from multiple causal variants at a locus. We developed a computational method to infer the probability of AH and applied it to three GWASs and four expression quantitative trait loci (eQTL) datasets. We identified a total of 4,152 loci with strong evidence of AH. The proportion of all loci with identified AH is 4%-23% in eQTLs, 35% in GWASs of high-density lipoprotein (HDL), and 23% in GWASs of schizophrenia. For eQTLs, we observed a strong correlation between sample size and the proportion of loci with AH (R2 = 0.85, p = 2.2 × 10-16), indicating that statistical power prevents identification of AH in other loci. Understanding the extent of AH may guide the development of new methods for fine mapping and association mapping of complex traits.
Collapse
Affiliation(s)
- Farhad Hormozdiari
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Anthony Zhu
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA
| | - Gleb Kichaev
- Bioinformatics IDP, University of California, Los Angeles, CA 90095, USA
| | - Chelsea J-T Ju
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA
| | - Ayellet V Segrè
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Jong Wha J Joo
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA; Department of Computer Science Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Hyejung Won
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Sriram Sankararaman
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Bogdan Pasaniuc
- Department of Human Genetics, University of California, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Sagiv Shifman
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
171
|
Wen X, Pique-Regi R, Luca F. Integrating molecular QTL data into genome-wide genetic association analysis: Probabilistic assessment of enrichment and colocalization. PLoS Genet 2017; 13:e1006646. [PMID: 28278150 PMCID: PMC5363995 DOI: 10.1371/journal.pgen.1006646] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 03/23/2017] [Accepted: 02/21/2017] [Indexed: 01/25/2023] Open
Abstract
We propose a novel statistical framework for integrating the result from molecular quantitative trait loci (QTL) mapping into genome-wide genetic association analysis of complex traits, with the primary objectives of quantitatively assessing the enrichment of the molecular QTLs in complex trait-associated genetic variants and the colocalizations of the two types of association signals. We introduce a natural Bayesian hierarchical model that treats the latent association status of molecular QTLs as SNP-level annotations for candidate SNPs of complex traits. We detail a computational procedure to seamlessly perform enrichment, fine-mapping and colocalization analyses, which is a distinct feature compared to the existing colocalization analysis procedures in the literature. The proposed approach is computationally efficient and requires only summary-level statistics. We evaluate and demonstrate the proposed computational approach through extensive simulation studies and analyses of blood lipid data and the whole blood eQTL data from the GTEx project. In addition, a useful utility from our proposed method enables the computation of expected colocalization signals using simple characteristics of the association data. Using this utility, we further illustrate the importance of enrichment analysis on the ability to discover colocalized signals and the potential limitations of currently available molecular QTL data. The software pipeline that implements the proposed computation procedures, enloc, is freely available at https://github.com/xqwen/integrative. Genome-wide association studies (GWAS) have been tremendously successful in identifying genetic variants that impact complex diseases. However, the roles of such studies in disease etiology remain poorly understood, primarily because a large proportion of the GWAS findings are located in the non-coding region of the genome. Recent advancements in high-throughput sequencing technology enable the systematic investigation of molecular quantitative trait loci (QTLs), which are genetic variants that directly affect molecular phenotypes (e.g., gene expression, transcription factor binding and DNA methylation). Linking molecular QTLs to GWAS findings intuitively represents an important step for interpreting the biological and clinical relevance of the GWAS results. In this paper, we describe a rigorous and efficient computational approach that assesses the enrichment and overlap between the GWAS findings and molecular QTLs. Importantly, we illustrate that the accurate quantification of overlapping between molecular QTL and GWAS signals requires reliable enrichment estimation. Our proposed approach fully accounts for the intrinsic uncertainty embedded in the association analyses of GWAS and molecular QTL mapping, and it outperforms the existing state-of-the-art approaches. Applying the proposed approach to the GWAS data of blood lipid traits and the whole blood expression QTLs (eQTLs) yields some novel biological insights and also illustrates the potential limitations of the currently available molecular QTL data.
Collapse
Affiliation(s)
- Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, United States of America
| |
Collapse
|
172
|
Mancuso N, Shi H, Goddard P, Kichaev G, Gusev A, Pasaniuc B. Integrating Gene Expression with Summary Association Statistics to Identify Genes Associated with 30 Complex Traits. Am J Hum Genet 2017; 100:473-487. [PMID: 28238358 PMCID: PMC5339290 DOI: 10.1016/j.ajhg.2017.01.031] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/23/2017] [Indexed: 01/24/2023] Open
Abstract
Although genome-wide association studies (GWASs) have identified thousands of risk loci for many complex traits and diseases, the causal variants and genes at these loci remain largely unknown. Here, we introduce a method for estimating the local genetic correlation between gene expression and a complex trait and utilize it to estimate the genetic correlation due to predicted expression between pairs of traits. We integrated gene expression measurements from 45 expression panels with summary GWAS data to perform 30 multi-tissue transcriptome-wide association studies (TWASs). We identified 1,196 genes whose expression is associated with these traits; of these, 168 reside more than 0.5 Mb away from any previously reported GWAS significant variant. We then used our approach to find 43 pairs of traits with significant genetic correlation at the level of predicted expression; of these, eight were not found through genetic correlation at the SNP level. Finally, we used bi-directional regression to find evidence that BMI causally influences triglyceride levels and that triglyceride levels causally influence low-density lipoprotein. Together, our results provide insight into the role of gene expression in the susceptibility of complex traits and diseases.
Collapse
Affiliation(s)
- Nicholas Mancuso
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA.
| | - Huwenbo Shi
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Pagé Goddard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Gleb Kichaev
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Alexander Gusev
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bogdan Pasaniuc
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90024, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA.
| |
Collapse
|
173
|
Limited statistical evidence for shared genetic effects of eQTLs and autoimmune-disease-associated loci in three major immune-cell types. Nat Genet 2017; 49:600-605. [PMID: 28218759 PMCID: PMC5374036 DOI: 10.1038/ng.3795] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/26/2017] [Indexed: 12/13/2022]
|
174
|
Pasaniuc B, Price AL. Dissecting the genetics of complex traits using summary association statistics. Nat Rev Genet 2017; 18:117-127. [PMID: 27840428 PMCID: PMC5449190 DOI: 10.1038/nrg.2016.142] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the past decade, genome-wide association studies (GWAS) have been used to successfully identify tens of thousands of genetic variants associated with complex traits and diseases. These studies have produced extensive repositories of genetic variation and trait measurements across large numbers of individuals, providing tremendous opportunities for further analyses. However, privacy concerns and other logistical considerations often limit access to individual-level genetic data, motivating the development of methods that analyse summary association statistics. Here, we review recent progress on statistical methods that leverage summary association data to gain insights into the genetic basis of complex traits and diseases.
Collapse
Affiliation(s)
- Bogdan Pasaniuc
- Departments of Human Genetics, and Pathology and Laboratory Medicine, University of California, Los Angeles, California 90095, USA
| | - Alkes L Price
- Departments of Epidemiology and Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
175
|
Kalmbach DA, Schneider LD, Cheung J, Bertrand SJ, Kariharan T, Pack AI, Gehrman PR. Genetic Basis of Chronotype in Humans: Insights From Three Landmark GWAS. Sleep 2017; 40:2662182. [PMID: 28364486 PMCID: PMC6084759 DOI: 10.1093/sleep/zsw048] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2016] [Indexed: 01/22/2023] Open
Abstract
Study Objectives Chronotype, or diurnal preference, refers to behavioral manifestations of the endogenous circadian system that governs preferred timing of sleep and wake. As variations in circadian timing and system perturbations are linked to disease development, the fundamental biology of chronotype has received attention for its role in the regulation and dysregulation of sleep and related illnesses. Family studies indicate that chronotype is a heritable trait, thus directing attention toward its genetic basis. Although discoveries from molecular studies of candidate genes have shed light onto its genetic architecture, the contribution of genetic variation to chronotype has remained unclear with few related variants identified. In the advent of large-scale genome-wide association studies (GWAS), scientists now have the ability to discover novel common genetic variants associated with complex phenotypes. Three recent large-scale GWASs of chronotype were conducted on subjects of European ancestry from the 23andMe cohort and the UK Biobank. This review discusses the findings of these landmark GWASs in the context of prior research. Methods We systematically reviewed and compared methodological and analytical approaches and results across the three GWASs of chronotype. Results A good deal of consistency was observed across studies with 9 genes identified in 2 of the 3 GWASs. Several genes previously unknown to influence chronotype were identified. Conclusions GWAS is an important tool in identifying common variants associated with the complex chronotype phenotype, the findings of which can supplement and guide molecular science. Future directions in model systems and discovery of rare variants are discussed.
Collapse
Affiliation(s)
- David A Kalmbach
- Departments of Psychiatry and Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Logan D Schneider
- Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94063
| | - Joseph Cheung
- Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94063
| | - Sarah J Bertrand
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital School of Medicine, Baltimore, MD 21205
| | - Thiruchelvam Kariharan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Allan I Pack
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104
| | - Philip R Gehrman
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
176
|
Lotta LA, Gulati P, Day FR, Payne F, Ongen H, van de Bunt M, Gaulton KJ, Eicher JD, Sharp SJ, Luan J, De Lucia Rolfe E, Stewart ID, Wheeler E, Willems SM, Adams C, Yaghootkar H, Forouhi NG, Khaw KT, Johnson AD, Semple RK, Frayling T, Perry JRB, Dermitzakis E, McCarthy MI, Barroso I, Wareham NJ, Savage DB, Langenberg C, O’Rahilly S, Scott RA. Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance. Nat Genet 2017; 49:17-26. [PMID: 27841877 PMCID: PMC5774584 DOI: 10.1038/ng.3714] [Citation(s) in RCA: 414] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023]
Abstract
Insulin resistance is a key mediator of obesity-related cardiometabolic disease, yet the mechanisms underlying this link remain obscure. Using an integrative genomic approach, we identify 53 genomic regions associated with insulin resistance phenotypes (higher fasting insulin levels adjusted for BMI, lower HDL cholesterol levels and higher triglyceride levels) and provide evidence that their link with higher cardiometabolic risk is underpinned by an association with lower adipose mass in peripheral compartments. Using these 53 loci, we show a polygenic contribution to familial partial lipodystrophy type 1, a severe form of insulin resistance, and highlight shared molecular mechanisms in common/mild and rare/severe insulin resistance. Population-level genetic analyses combined with experiments in cellular models implicate CCDC92, DNAH10 and L3MBTL3 as previously unrecognized molecules influencing adipocyte differentiation. Our findings support the notion that limited storage capacity of peripheral adipose tissue is an important etiological component in insulin-resistant cardiometabolic disease and highlight genes and mechanisms underpinning this link.
Collapse
Affiliation(s)
- Luca A. Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Pawan Gulati
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Felix R. Day
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Felicity Payne
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United
Kingdom
| | - Halit Ongen
- Department of Genetic Medicine and Development, University of Geneva
Medical School, Geneva, Switzerland
| | - Martijn van de Bunt
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University
of Oxford, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford,
Oxford, United Kingdom
| | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La
Jolla, USA
| | - John D. Eicher
- Population Sciences Branch, Division of Intramural Research,
National Heart, Lung and Blood Institute, Bethesda, USA
| | - Stephen J. Sharp
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Jian’an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | | | - Isobel D. Stewart
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Eleanor Wheeler
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United
Kingdom
| | - Sara M. Willems
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Claire Adams
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, Institute of Biomedical and Clinical
Science, University of Exeter Medical School, Royal Devon and Exeter Hospital,
Exeter, United Kingdom
| | | | | | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of
Cambridge, Cambridge, United Kingdom
| | - Andrew D. Johnson
- Population Sciences Branch, Division of Intramural Research,
National Heart, Lung and Blood Institute, Bethesda, USA
| | - Robert K. Semple
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Timothy Frayling
- Genetics of Complex Traits, Institute of Biomedical and Clinical
Science, University of Exeter Medical School, Royal Devon and Exeter Hospital,
Exeter, United Kingdom
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Emmanouil Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva
Medical School, Geneva, Switzerland
| | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University
of Oxford, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford,
Oxford, United Kingdom
| | - Inês Barroso
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United
Kingdom
| | | | - David B. Savage
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| | - Stephen O’Rahilly
- Metabolic Research Laboratories, Institute of Metabolic Science,
University of Cambridge, Cambridge, United Kingdom
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United
Kingdom
| |
Collapse
|
177
|
Deplancke B, Alpern D, Gardeux V. The Genetics of Transcription Factor DNA Binding Variation. Cell 2016; 166:538-554. [PMID: 27471964 DOI: 10.1016/j.cell.2016.07.012] [Citation(s) in RCA: 267] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Indexed: 12/23/2022]
Abstract
Most complex trait-associated variants are located in non-coding regulatory regions of the genome, where they have been shown to disrupt transcription factor (TF)-DNA binding motifs. Variable TF-DNA interactions are therefore increasingly considered as key drivers of phenotypic variation. However, recent genome-wide studies revealed that the majority of variable TF-DNA binding events are not driven by sequence alterations in the motif of the studied TF. This observation implies that the molecular mechanisms underlying TF-DNA binding variation and, by extrapolation, inter-individual phenotypic variation are more complex than originally anticipated. Here, we summarize the findings that led to this important paradigm shift and review proposed mechanisms for local, proximal, or distal genetic variation-driven variable TF-DNA binding. In addition, we discuss the biomedical implications of these findings for our ability to dissect the molecular role(s) of non-coding genetic variants in complex traits, including disease susceptibility.
Collapse
Affiliation(s)
- Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Vincent Gardeux
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| |
Collapse
|
178
|
Hormozdiari F, van de Bunt M, Segrè AV, Li X, Joo JWJ, Bilow M, Sul JH, Sankararaman S, Pasaniuc B, Eskin E. Colocalization of GWAS and eQTL Signals Detects Target Genes. Am J Hum Genet 2016; 99:1245-1260. [PMID: 27866706 DOI: 10.1016/j.ajhg.2016.10.003] [Citation(s) in RCA: 470] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/03/2016] [Indexed: 01/01/2023] Open
Abstract
The vast majority of genome-wide association study (GWAS) risk loci fall in non-coding regions of the genome. One possible hypothesis is that these GWAS risk loci alter the individual's disease risk through their effect on gene expression in different tissues. In order to understand the mechanisms driving a GWAS risk locus, it is helpful to determine which gene is affected in specific tissue types. For example, the relevant gene and tissue could play a role in the disease mechanism if the same variant responsible for a GWAS locus also affects gene expression. Identifying whether or not the same variant is causal in both GWASs and expression quantitative trail locus (eQTL) studies is challenging because of the uncertainty induced by linkage disequilibrium and the fact that some loci harbor multiple causal variants. However, current methods that address this problem assume that each locus contains a single causal variant. In this paper, we present eCAVIAR, a probabilistic method that has several key advantages over existing methods. First, our method can account for more than one causal variant in any given locus. Second, it can leverage summary statistics without accessing the individual genotype data. We use both simulated and real datasets to demonstrate the utility of our method. Using publicly available eQTL data on 45 different tissues, we demonstrate that eCAVIAR can prioritize likely relevant tissues and target genes for a set of glucose- and insulin-related trait loci.
Collapse
Affiliation(s)
- Farhad Hormozdiari
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Martijn van de Bunt
- Oxford Centre for Diabetes, Endocrinology, & Metabolism, University of Oxford, Oxford OX3 7LJ, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Ayellet V Segrè
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiao Li
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jong Wha J Joo
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Bilow
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jae Hoon Sul
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA; Semel Center for Informatics and Personalized Genomics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sriram Sankararaman
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
179
|
Bastami M, Nariman-Saleh-Fam Z, Saadatian Z, Nariman-Saleh-Fam L, Omrani MD, Ghaderian SMH, Masotti A. The miRNA targetome of coronary artery disease is perturbed by functional polymorphisms identified and prioritized by in-depth bioinformatics analyses exploiting genome-wide association studies. Gene 2016; 594:74-81. [DOI: 10.1016/j.gene.2016.08.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/27/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022]
|
180
|
Groop L. New approaches beyond genetics: towards precision medicine in diabetes. Diabetologia 2016; 59:2495-2496. [PMID: 27722776 DOI: 10.1007/s00125-016-4014-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 04/27/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Leif Groop
- Lund University Diabetes Centre, Jan Waldenströmsgata 35, 20505, Malmö, Sweden.
| |
Collapse
|
181
|
Wheeler HE, Shah KP, Brenner J, Garcia T, Aquino-Michaels K, Cox NJ, Nicolae DL, Im HK. Survey of the Heritability and Sparse Architecture of Gene Expression Traits across Human Tissues. PLoS Genet 2016; 12:e1006423. [PMID: 27835642 PMCID: PMC5106030 DOI: 10.1371/journal.pgen.1006423] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 10/12/2016] [Indexed: 11/19/2022] Open
Abstract
Understanding the genetic architecture of gene expression traits is key to elucidating the underlying mechanisms of complex traits. Here, for the first time, we perform a systematic survey of the heritability and the distribution of effect sizes across all representative tissues in the human body. We find that local h2 can be relatively well characterized with 59% of expressed genes showing significant h2 (FDR < 0.1) in the DGN whole blood cohort. However, current sample sizes (n ≤ 922) do not allow us to compute distal h2. Bayesian Sparse Linear Mixed Model (BSLMM) analysis provides strong evidence that the genetic contribution to local expression traits is dominated by a handful of genetic variants rather than by the collective contribution of a large number of variants each of modest size. In other words, the local architecture of gene expression traits is sparse rather than polygenic across all 40 tissues (from DGN and GTEx) examined. This result is confirmed by the sparsity of optimal performing gene expression predictors via elastic net modeling. To further explore the tissue context specificity, we decompose the expression traits into cross-tissue and tissue-specific components using a novel Orthogonal Tissue Decomposition (OTD) approach. Through a series of simulations we show that the cross-tissue and tissue-specific components are identifiable via OTD. Heritability and sparsity estimates of these derived expression phenotypes show similar characteristics to the original traits. Consistent properties relative to prior GTEx multi-tissue analysis results suggest that these traits reflect the expected biology. Finally, we apply this knowledge to develop prediction models of gene expression traits for all tissues. The prediction models, heritability, and prediction performance R2 for original and decomposed expression phenotypes are made publicly available (https://github.com/hakyimlab/PrediXcan).
Collapse
Affiliation(s)
- Heather E. Wheeler
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
- Department of Computer Science, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Kaanan P. Shah
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Jonathon Brenner
- Department of Computer Science, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Tzintzuni Garcia
- Center for Research Informatics, University of Chicago, Chicago, Illinois, United States of America
| | - Keston Aquino-Michaels
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | | | - Nancy J. Cox
- Division of Genetic Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Dan L. Nicolae
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Hae Kyung Im
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
182
|
Glastonbury C, Viñuela A, Buil A, Halldorsson G, Thorleifsson G, Helgason H, Thorsteinsdottir U, Stefansson K, Dermitzakis E, Spector T, Small K. Adiposity-Dependent Regulatory Effects on Multi-tissue Transcriptomes. Am J Hum Genet 2016; 99:567-579. [PMID: 27588447 PMCID: PMC5011064 DOI: 10.1016/j.ajhg.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/01/2016] [Indexed: 10/25/2022] Open
Abstract
Obesity is a global epidemic that is causally associated with a range of diseases, including type 2 diabetes and cardiovascular disease, at the population-level. However, there is marked heterogeneity in obesity-related outcomes among individuals. This might reflect genotype-dependent responses to adiposity. Given that adiposity, measured by BMI, is associated with widespread changes in gene expression and regulatory variants mediate the majority of known complex trait loci, we sought to identify gene-by-BMI (G × BMI) interactions on the regulation of gene expression in a multi-tissue RNA-sequencing (RNA-seq) dataset from the TwinsUK cohort (n = 856). At a false discovery rate of 5%, we identified 16 cis G × BMI interactions (top cis interaction: CHURC1, rs7143432, p = 2.0 × 10(-12)) and one variant regulating 53 genes in trans (top trans interaction: ZNF423, rs3851570, p = 8.2 × 10(-13)), all in adipose tissue. The interactions were adipose-specific and enriched for variants overlapping adipocyte enhancers, and regulated genes were enriched for metabolic and inflammatory processes. We replicated a subset of the interactions in an independent adipose RNA-seq dataset (deCODE genetics, n = 754). We also confirmed the interactions with an alternate measure of obesity, dual-energy X-ray absorptiometry (DXA)-derived visceral-fat-volume measurements, in a subset of TwinsUK individuals (n = 682). The identified G × BMI regulatory effects demonstrate the dynamic nature of gene regulation and reveal a functional mechanism underlying the heterogeneous response to obesity. Additionally, we have provided a web browser allowing interactive exploration of the dataset, including of association between expression, BMI, and G × BMI regulatory effects in four tissues.
Collapse
|
183
|
RNA Sequencing and Genetic Disease. CURRENT GENETIC MEDICINE REPORTS 2016. [DOI: 10.1007/s40142-016-0098-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
184
|
Franzén O, Ermel R, Cohain A, Akers NK, Di Narzo A, Talukdar HA, Foroughi-Asl H, Giambartolomei C, Fullard JF, Sukhavasi K, Köks S, Gan LM, Giannarelli C, Kovacic JC, Betsholtz C, Losic B, Michoel T, Hao K, Roussos P, Skogsberg J, Ruusalepp A, Schadt EE, Björkegren JLM. Cardiometabolic risk loci share downstream cis- and trans-gene regulation across tissues and diseases. Science 2016; 353:827-30. [PMID: 27540175 DOI: 10.1126/science.aad6970] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 07/22/2016] [Indexed: 12/11/2022]
Abstract
Genome-wide association studies (GWAS) have identified hundreds of cardiometabolic disease (CMD) risk loci. However, they contribute little to genetic variance, and most downstream gene-regulatory mechanisms are unknown. We genotyped and RNA-sequenced vascular and metabolic tissues from 600 coronary artery disease patients in the Stockholm-Tartu Atherosclerosis Reverse Networks Engineering Task study (STARNET). Gene expression traits associated with CMD risk single-nucleotide polymorphism (SNPs) identified by GWAS were more extensively found in STARNET than in tissue- and disease-unspecific gene-tissue expression studies, indicating sharing of downstream cis-/trans-gene regulation across tissues and CMDs. In contrast, the regulatory effects of other GWAS risk SNPs were tissue-specific; abdominal fat emerged as an important gene-regulatory site for blood lipids, such as for the low-density lipoprotein cholesterol and coronary artery disease risk gene PCSK9 STARNET provides insights into gene-regulatory mechanisms for CMD risk loci, facilitating their translation into opportunities for diagnosis, therapy, and prevention.
Collapse
Affiliation(s)
- Oscar Franzén
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Clinical Gene Networks AB, Jungfrugatan 10, 114 44 Stockholm, Sweden
| | - Raili Ermel
- Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia. Department of Cardiac Surgery, Tartu University Hospital, 1a Ludwig Puusepa Street, 50406 Tartu, Estonia
| | - Ariella Cohain
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Nicholas K Akers
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Antonio Di Narzo
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Husain A Talukdar
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden
| | - Hassan Foroughi-Asl
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden
| | - Claudia Giambartolomei
- Division of Psychiatric Genomics, Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - John F Fullard
- Division of Psychiatric Genomics, Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Katyayani Sukhavasi
- Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia
| | - Sulev Köks
- Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia
| | - Li-Ming Gan
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Chiara Giannarelli
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Cardiovascular Research Center Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Jason C Kovacic
- Cardiovascular Research Center Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Christer Betsholtz
- AstraZeneca-Karolinska Integrated CardioMetabolic Centre (ICMC), Karolinska Institutet, Novum, Blickagången 6, 141 57 Huddinge, Sweden. Department of Immunology, Genetics and Pathology Dag Hammarskjölds Väg 20, 751 85 Uppsala, Sweden
| | - Bojan Losic
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Tom Michoel
- Division of Genetics and Genomics, The Roslin Institute, University of Edinburgh, Old College, South Bridge, Edinburgh EH8 9YL, UK
| | - Ke Hao
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Division of Psychiatric Genomics, Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Department of Psychiatry, J. J. Peters VA Medical Center, Mental Illness Research Education and Clinical Center (MIRECC), 130 West Kingsbridge Road, Bronx, NY 10468, USA
| | - Josefin Skogsberg
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden
| | - Arno Ruusalepp
- Clinical Gene Networks AB, Jungfrugatan 10, 114 44 Stockholm, Sweden. Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia. Department of Cardiac Surgery, Tartu University Hospital, 1a Ludwig Puusepa Street, 50406 Tartu, Estonia
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Clinical Gene Networks AB, Jungfrugatan 10, 114 44 Stockholm, Sweden. Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia. Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden.
| |
Collapse
|
185
|
Peterson CB, Bogomolov M, Benjamini Y, Sabatti C. TreeQTL: hierarchical error control for eQTL findings. Bioinformatics 2016; 32:2556-8. [PMID: 27153635 PMCID: PMC4978936 DOI: 10.1093/bioinformatics/btw198] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 03/25/2016] [Accepted: 04/10/2016] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED : Commonly used multiplicity adjustments fail to control the error rate for reported findings in many expression quantitative trait loci (eQTL) studies. TreeQTL implements a hierarchical multiple testing procedure which allows control of appropriate error rates defined relative to a grouping of the eQTL hypotheses. AVAILABILITY AND IMPLEMENTATION The R package TreeQTL is available for download at http://bioinformatics.org/treeqtl CONTACT sabatti@stanford.edu SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- C B Peterson
- Department of Health Research and Policy, Stanford University, Stanford, CA 94305, USA
| | - M Bogomolov
- Faculty of Industrial Engineering and Management, Technion, Haifa 32000, Israel
| | - Y Benjamini
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv 6997801, Israel
| | - C Sabatti
- Departments of Biomedical Data Science and Statistics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
186
|
Morris DL, Sheng Y, Zhang Y, Wang YF, Zhu Z, Tombleson P, Chen L, Cunninghame Graham DS, Bentham J, Roberts AL, Chen R, Zuo X, Wang T, Wen L, Yang C, Liu L, Yang L, Li F, Huang Y, Yin X, Yang S, Rönnblom L, Fürnrohr BG, Voll RE, Schett G, Costedoat-Chalumeau N, Gaffney PM, Lau YL, Zhang X, Yang W, Cui Y, Vyse TJ. Genome-wide association meta-analysis in Chinese and European individuals identifies ten new loci associated with systemic lupus erythematosus. Nat Genet 2016; 48:940-946. [PMID: 27399966 PMCID: PMC4966635 DOI: 10.1038/ng.3603] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/01/2016] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE; OMIM 152700) is a genetically complex autoimmune disease. Genome-wide association studies (GWASs) have identified more than 50 loci as robustly associated with the disease in single ancestries, but genome-wide transancestral studies have not been conducted. We combined three GWAS data sets from Chinese (1,659 cases and 3,398 controls) and European (4,036 cases and 6,959 controls) populations. A meta-analysis of these studies showed that over half of the published SLE genetic associations are present in both populations. A replication study in Chinese (3,043 cases and 5,074 controls) and European (2,643 cases and 9,032 controls) subjects found ten previously unreported SLE loci. Our study provides further evidence that the majority of genetic risk polymorphisms for SLE are contained within the same regions across both populations. Furthermore, a comparison of risk allele frequencies and genetic risk scores suggested that the increased prevalence of SLE in non-Europeans (including Asians) has a genetic basis.
Collapse
Affiliation(s)
- David L Morris
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Yujun Sheng
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Yan Zhang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yong-Fei Wang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Zhengwei Zhu
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Philip Tombleson
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Lingyan Chen
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | | | - James Bentham
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Amy L Roberts
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Ruoyan Chen
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xianbo Zuo
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Tingyou Wang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Leilei Wen
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Chao Yang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Lu Liu
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Lulu Yang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Feng Li
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Yuanbo Huang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Xianyong Yin
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Sen Yang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Lars Rönnblom
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Barbara G Fürnrohr
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
- Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
- Division of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, Austria
- Division of Biological Chemistry, Medical University Innsbruck, Innsbruck, Austria
| | - Reinhard E Voll
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
- Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
- Department of Rheumatology, University Hospital Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, University Hospital Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency, University Hospital Freiburg, Freiburg, Germany
| | - Georg Schett
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
- Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nathalie Costedoat-Chalumeau
- AP-HP, Hôpital Cochin, Centre de référence maladies auto-immunes et systémiques rares, Paris, France
- Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Xuejun Zhang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
- Department of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yong Cui
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Timothy J Vyse
- Division of Genetics and Molecular Medicine, King's College London, London, UK
- Division of Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| |
Collapse
|
187
|
Lappalainen T. Functional genomics bridges the gap between quantitative genetics and molecular biology. Genome Res 2016; 25:1427-31. [PMID: 26430152 PMCID: PMC4579327 DOI: 10.1101/gr.190983.115] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Deep characterization of molecular function of genetic variants in the human genome is becoming increasingly important for understanding genetic associations to disease and for learning to read the regulatory code of the genome. In this paper, I discuss how recent advances in both quantitative genetics and molecular biology have contributed to understanding functional effects of genetic variants, lessons learned from eQTL studies, and future challenges in this field.
Collapse
Affiliation(s)
- Tuuli Lappalainen
- New York Genome Center, New York, New York 10013, USA; Department of Systems Biology, Columbia University, New York, New York 10032, USA
| |
Collapse
|
188
|
Abstract
There are thousands of known associations between genetic variants and complex human phenotypes, and the rate of novel discoveries is rapidly increasing. Translating those associations into knowledge of disease mechanisms remains a fundamental challenge because the associated variants are overwhelmingly in noncoding regions of the genome where we have few guiding principles to predict their function. Intersecting the compendium of identified genetic associations with maps of regulatory activity across the human genome has revealed that phenotype-associated variants are highly enriched in candidate regulatory elements. Allele-specific analyses of gene regulation can further prioritize variants that likely have a functional effect on disease mechanisms; and emerging high-throughput assays to quantify the activity of candidate regulatory elements are a promising next step in that direction. Together, these technologies have created the ability to systematically and empirically test hypotheses about the function of noncoding variants and haplotypes at the scale needed for comprehensive and systematic follow-up of genetic association studies. Major coordinated efforts to quantify regulatory mechanisms across genetically diverse populations in increasingly realistic cell models would be highly beneficial to realize that potential.
Collapse
Affiliation(s)
- William L Lowe
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Timothy E Reddy
- Department of Biostatistics and Bioinformatics, Duke University Medical School, Durham, North Carolina 27708, USA; Center for Genomic and Computational Biology, Duke University Medical School, Durham, North Carolina 27708, USA
| |
Collapse
|
189
|
Abstract
BACKGROUND Prepulse inhibition (PPI) of the startle reflex has been suggested as a candidate endophenotype for schizophrenia research, as it shows high heritability and has been found deficient in schizophrenia spectrum disorders. The objectives of the study were to 1) identify common genetic variants associated with baseline startle and PPI; 2) estimate the single nucleotide polymorphism heritability; and 3) examine the relationship of polygenic score for schizophrenia with baseline startle and PPI. METHODS A cohort of healthy young male subjects (n = 1493) originating from the Learning on Genetics of Schizophrenia Spectrum project was assessed for baseline startle and PPI. The most recent genome-wide association study in schizophrenia from the Psychiatric Genomics Consortium 2 was used to calculate polygenic scores. RESULTS Eleven loci showed suggestive association (p < 10(-6)) with baseline startle and PPI in the discovery cohort. Additional genotyping in a replication cohort identified genome-wide significant association at two loci (rs61810702 and rs4718984). These loci were co-localized with expression quantitative trait loci associated with gene expression of nerve growth factor (NGF) and calneuron 1 (CALN1) genes. Estimation of the genetic and environmental contributions to baseline startle and PPI showed a substantial single nucleotide polymorphism heritability for 120-ms PPI stimuli. Increased polygenic risk score for schizophrenia was associated with reduced PPI. CONCLUSIONS Common genetic variation has an important role in the etiology of schizophrenia and PPI impairments. Overall, these data support the idea that PPI is a valid endophenotype that can be used to explore the genetic architecture of schizophrenia.
Collapse
|
190
|
Yarwood A, Eyre S, Worthington J. Genetic susceptibility to rheumatoid arthritis and its implications for novel drug discovery. Expert Opin Drug Discov 2016; 11:805-13. [PMID: 27267163 DOI: 10.1080/17460441.2016.1195366] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Over 100 susceptibility loci have now been identified for rheumatoid arthritis (RA), several of which are already the targets of approved RA therapies providing proof of concept for the use of genetics in novel drug development for RA. Determining how these loci contribute to disease will be key to elucidating the mechanisms driving disease development, which has the potential for major impact on therapeutic development. AREAS COVERED Here the authors review the use of genetics in drug discovery, including the use of 'omics' data to prioritise potential drug targets at susceptibility loci using RA as an exemplar. They discuss the current state of RA genetics its impact on stratified medicine, and how the findings from RA genetics studies can be used to inform drug discovery. EXPERT OPINION It is anticipated that functional characterisation of disease variants will provide biological validation of a gene as a drug target, providing safer targets, with an increased likelihood of efficacy. In the future, techniques such as genome editing may represent a plausible option for RA therapy. Technologies such as genome-wide chromatin conformation capture Hi-C and CRISPR will be crucial to inform our understanding of how diseases develop and in developing new treatments.
Collapse
Affiliation(s)
- Annie Yarwood
- a Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre , The University of Manchester , Manchester , UK
| | - Steve Eyre
- a Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre , The University of Manchester , Manchester , UK
| | - Jane Worthington
- a Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre , The University of Manchester , Manchester , UK.,b NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust , Manchester Academic Health Science Centre , Manchester , UK
| |
Collapse
|
191
|
Peterson CB, Service SK, Jasinska AJ, Gao F, Zelaya I, Teshiba TM, Bearden CE, Cantor RM, Reus VI, Macaya G, López-Jaramillo C, Bogomolov M, Benjamini Y, Eskin E, Coppola G, Freimer NB, Sabatti C. Characterization of Expression Quantitative Trait Loci in Pedigrees from Colombia and Costa Rica Ascertained for Bipolar Disorder. PLoS Genet 2016; 12:e1006046. [PMID: 27176483 PMCID: PMC4866754 DOI: 10.1371/journal.pgen.1006046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/20/2016] [Indexed: 01/22/2023] Open
Abstract
The observation that variants regulating gene expression (expression quantitative trait loci, eQTL) are at a high frequency among SNPs associated with complex traits has made the genome-wide characterization of gene expression an important tool in genetic mapping studies of such traits. As part of a study to identify genetic loci contributing to bipolar disorder and other quantitative traits in members of 26 pedigrees from Costa Rica and Colombia, we measured gene expression in lymphoblastoid cell lines derived from 786 pedigree members. The study design enabled us to comprehensively reconstruct the genetic regulatory network in these families, provide estimates of heritability, identify eQTL, evaluate missing heritability for the eQTL, and quantify the number of different alleles contributing to any given locus. In the eQTL analysis, we utilize a recently proposed hierarchical multiple testing strategy which controls error rates regarding the discovery of functional variants. Our results elucidate the heritability and regulation of gene expression in this unique Latin American study population and identify a set of regulatory SNPs which may be relevant in future investigations of complex disease in this population. Since our subjects belong to extended families, we are able to compare traditional kinship-based estimates with those from more recent methods that depend only on genotype information.
Collapse
Affiliation(s)
- Christine B. Peterson
- Department of Health Research and Policy, Stanford University, Stanford, California, United States of America
| | - Susan K. Service
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Anna J. Jasinska
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Fuying Gao
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
| | - Ivette Zelaya
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
| | - Terri M. Teshiba
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Rita M. Cantor
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Victor I. Reus
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
| | - Gabriel Macaya
- Cell and Molecular Biology Research Center, Universidad de Costa Rica, San Pedro de Montes de Oca, San José, Costa Rica
| | - Carlos López-Jaramillo
- Grupo de Investigación en Psiquiatría (Research Group in Psychiatry (GIPSI)), Departamento de Psiquiatría, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia; Mood Disorders Program, Hospital San Vicente Fundacion, Medellín, Colombia
| | - Marina Bogomolov
- Faculty of Industrial Engineering and Management, Technion, Haifa, Israel
| | - Yoav Benjamini
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
| | - Eleazar Eskin
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Computer Science, University of California Los Angeles, Los Angeles, California, United States of America
| | - Giovanni Coppola
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Nelson B. Freimer
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Chiara Sabatti
- Department of Biomedical Data Science and Department of Statistics, Stanford University, Stanford, California, United States of America
| |
Collapse
|
192
|
Integrative genomics analyses unveil downstream biological effectors of disease-specific polymorphisms buried in intergenic regions. NPJ Genom Med 2016; 1. [PMID: 27482468 PMCID: PMC4966659 DOI: 10.1038/npjgenmed.2016.6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Functionally altered biological mechanisms arising from disease-associated polymorphisms, remain difficult to characterise when those variants are intergenic, or, fall between genes. We sought to identify shared downstream mechanisms by which inter- and intragenic single-nucleotide polymorphisms (SNPs) contribute to a specific physiopathology. Using computational modelling of 2 million pairs of disease-associated SNPs drawn from genome-wide association studies (GWAS), integrated with expression Quantitative Trait Loci (eQTL) and Gene Ontology functional annotations, we predicted 3,870 inter–intra and inter–intra SNP pairs with convergent biological mechanisms (FDR<0.05). These prioritised SNP pairs with overlapping messenger RNA targets or similar functional annotations were more likely to be associated with the same disease than unrelated pathologies (OR>12). We additionally confirmed synergistic and antagonistic genetic interactions for a subset of prioritised SNP pairs in independent studies of Alzheimer’s disease (entropy P=0.046), bladder cancer (entropy P=0.039), and rheumatoid arthritis (PheWAS case–control P<10−4). Using ENCODE data sets, we further statistically validated that the biological mechanisms shared within prioritised SNP pairs are frequently governed by matching transcription factor binding sites and long-range chromatin interactions. These results provide a ‘roadmap’ of disease mechanisms emerging from GWAS and further identify candidate therapeutic targets among downstream effectors of intergenic SNPs.
Collapse
|
193
|
Kukurba KR, Parsana P, Balliu B, Smith KS, Zappala Z, Knowles DA, Favé MJ, Davis JR, Li X, Zhu X, Potash JB, Weissman MM, Shi J, Kundaje A, Levinson DF, Awadalla P, Mostafavi S, Battle A, Montgomery SB. Impact of the X Chromosome and sex on regulatory variation. Genome Res 2016; 26:768-77. [PMID: 27197214 PMCID: PMC4889977 DOI: 10.1101/gr.197897.115] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 04/18/2016] [Indexed: 02/07/2023]
Abstract
The X Chromosome, with its unique mode of inheritance, contributes to differences between the sexes at a molecular level, including sex-specific gene expression and sex-specific impact of genetic variation. Improving our understanding of these differences offers to elucidate the molecular mechanisms underlying sex-specific traits and diseases. However, to date, most studies have either ignored the X Chromosome or had insufficient power to test for the sex-specific impact of genetic variation. By analyzing whole blood transcriptomes of 922 individuals, we have conducted the first large-scale, genome-wide analysis of the impact of both sex and genetic variation on patterns of gene expression, including comparison between the X Chromosome and autosomes. We identified a depletion of expression quantitative trait loci (eQTL) on the X Chromosome, especially among genes under high selective constraint. In contrast, we discovered an enrichment of sex-specific regulatory variants on the X Chromosome. To resolve the molecular mechanisms underlying such effects, we generated chromatin accessibility data through ATAC-sequencing to connect sex-specific chromatin accessibility to sex-specific patterns of expression and regulatory variation. As sex-specific regulatory variants discovered in our study can inform sex differences in heritable disease prevalence, we integrated our data with genome-wide association study data for multiple immune traits identifying several traits with significant sex biases in genetic susceptibilities. Together, our study provides genome-wide insight into how genetic variation, the X Chromosome, and sex shape human gene regulation and disease.
Collapse
Affiliation(s)
- Kimberly R Kukurba
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Princy Parsana
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Brunilda Balliu
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Kevin S Smith
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Zachary Zappala
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - David A Knowles
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Marie-Julie Favé
- Sainte-Justine University Hospital Research Centre, Department of Pediatrics, University of Montreal, Montreal, Québec H3T 1J4, Canada
| | - Joe R Davis
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Xin Li
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Xiaowei Zhu
- Department of Psychiatry, Stanford University School of Medicine, Stanford, California 94305, USA
| | - James B Potash
- Department of Psychiatry, University of Iowa Hospitals & Clinics, Iowa City, Iowa 52242, USA
| | - Myrna M Weissman
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, New York 10032, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA; Department of Computer Science, Stanford University, Stanford, California 94305, USA
| | - Douglas F Levinson
- Department of Psychiatry, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Philip Awadalla
- Sainte-Justine University Hospital Research Centre, Department of Pediatrics, University of Montreal, Montreal, Québec H3T 1J4, Canada
| | - Sara Mostafavi
- Department of Statistics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Alexis Battle
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA;
| | - Stephen B Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA; Department of Computer Science, Stanford University, Stanford, California 94305, USA;
| |
Collapse
|
194
|
Nakaoka H, Gurumurthy A, Hayano T, Ahmadloo S, Omer WH, Yoshihara K, Yamamoto A, Kurose K, Enomoto T, Akira S, Hosomichi K, Inoue I. Allelic Imbalance in Regulation of ANRIL through Chromatin Interaction at 9p21 Endometriosis Risk Locus. PLoS Genet 2016; 12:e1005893. [PMID: 27055116 PMCID: PMC4824487 DOI: 10.1371/journal.pgen.1005893] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/02/2016] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association studies (GWASs) have discovered numerous single nucleotide polymorphisms (SNPs) associated with human complex disorders. However, functional characterization of the disease-associated SNPs remains a formidable challenge. Here we explored regulatory mechanism of a SNP on chromosome 9p21 associated with endometriosis by leveraging “allele-specific” functional genomic approaches. By re-sequencing 1.29 Mb of 9p21 region and scrutinizing DNase-seq data from the ENCODE project, we prioritized rs17761446 as a candidate functional variant that was in perfect linkage disequilibrium with the original GWAS SNP (rs10965235) and located on DNase I hypersensitive site. Chromosome conformation capture followed by high-throughput sequencing revealed that the protective G allele of rs17761446 exerted stronger chromatin interaction with ANRIL promoter. We demonstrated that the protective allele exhibited preferential binding affinities to TCF7L2 and EP300 by bioinformatics and chromatin immunoprecipitation (ChIP) analyses. ChIP assays for histone H3 lysine 27 acetylation and RNA polymerase II reinforced the enhancer activity of the SNP site. The allele specific expression analysis for eutopic endometrial tissues and endometrial carcinoma cell lines showed that rs17761446 was a cis-regulatory variant where G allele was associated with increased ANRIL expression. Our work illuminates the allelic imbalances in a series of transcriptional regulation from factor binding to gene expression mediated by chromatin interaction underlie the molecular mechanism of 9p21 endometriosis risk locus. Functional genomics on common disease will unlock functional aspect of genotype-phenotype correlations in the post-GWAS stage. A large number of variants associated with human complex diseases have been discovered by genome-wide association studies (GWASs). These discoveries have been anticipated to be translated into the definitive understanding of disease pathogeneses; however, functional characterization of the disease-associated SNPs remains a formidable challenge. Here we explored regulatory mechanism of a variant on chromosome 9p21 associated with endometriosis, a common gynecological disorder. By scrutinizing linkage disequilibrium structure and DNase I hypersensitive sites across the risk locus, we prioritized rs17761446 as a candidate causal variant. The results of our “allele-specific” functional genomic approaches sheds light on regulatory mechanisms underlying 9p21 endometriosis risk locus, in which preferential bindings of TCF7L2 and its coactivator EP300 to the protective G allele of rs17761446 lead to stronger chromatin interaction with the promoter of ANRIL, which in turn activate transcription of the non-coding RNA. Motivated by the fact that TCF7L2 was a key transcription factor of Wnt signaling pathway, we postulated that the induction of Wnt signaling activated expression levels of ANRIL and cell cycle inhibitors, CDKN2A/2B. Functional genomics on common disease will unlock functional aspect of genotype-phenotype correlations in the post-GWAS stage.
Collapse
Affiliation(s)
- Hirofumi Nakaoka
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Aishwarya Gurumurthy
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Takahide Hayano
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Somayeh Ahmadloo
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Waleed H Omer
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, Japan
| | - Akihito Yamamoto
- Department of Obstetrics and Gynecology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Keisuke Kurose
- Department of Obstetrics and Gynecology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, Japan
| | - Shigeo Akira
- Department of Obstetrics and Gynecology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Kazuyoshi Hosomichi
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Ituro Inoue
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
- * E-mail:
| |
Collapse
|
195
|
Stracquadanio G, Wang X, Wallace M, Grawenda AM, Zhang P, Hewitt J, Zeron-Medina J, Castro-Giner F, Tomlinson IP, Goding CR, Cygan KJ, Fairbrother WG, Thomas LF, Sætrom P, Gemignani F, Landi S, Schuster-Boeckler B, Bell DA, Bond GL. The importance of p53 pathway genetics in inherited and somatic cancer genomes. Nat Rev Cancer 2016; 16:251-65. [PMID: 27009395 PMCID: PMC6854702 DOI: 10.1038/nrc.2016.15] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decades of research have shown that mutations in the p53 stress response pathway affect the incidence of diverse cancers more than mutations in other pathways. However, most evidence is limited to somatic mutations and rare inherited mutations. Using newly abundant genomic data, we demonstrate that commonly inherited genetic variants in the p53 pathway also affect the incidence of a broad range of cancers more than variants in other pathways. The cancer-associated single nucleotide polymorphisms (SNPs) of the p53 pathway have strikingly similar genetic characteristics to well-studied p53 pathway cancer-causing somatic mutations. Our results enable insights into p53-mediated tumour suppression in humans and into p53 pathway-based cancer surveillance and treatment strategies.
Collapse
Affiliation(s)
- Giovanni Stracquadanio
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Xuting Wang
- Environmental Genomics Group, Genome Integrity and Structural Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Marsha Wallace
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Anna M. Grawenda
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Ping Zhang
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Juliet Hewitt
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Jorge Zeron-Medina
- Vall d’Hebron University Hospital, Oncology Department, Passeig de la Vall D’Hebron 119, 08035 Barcelona, Spain
| | - Francesc Castro-Giner
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Ian P. Tomlinson
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Colin R. Goding
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Kamil J. Cygan
- Center for Computational Molecular Biology, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - William G. Fairbrother
- Center for Computational Molecular Biology, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Laurent F. Thomas
- Department of Cancer Research and Molecular Medicine, Norwegian, University of Science and Technology, NO-7491 Trondheim, Norway
| | - Pål Sætrom
- Department of Computer and Information Science, Norwegian, University of Science and Technology, NO-7491 Trondheim, Norway
- Department of Cancer Research and Molecular Medicine, Norwegian, University of Science and Technology, NO-7491 Trondheim, Norway
| | - Frederica Gemignani
- Genetics- Department of Biology, University of Pisa, Via Derna, 1, 56126 Pisa - Italy
| | - Stefano Landi
- Genetics- Department of Biology, University of Pisa, Via Derna, 1, 56126 Pisa - Italy
| | - Benjamin Schuster-Boeckler
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Douglas A. Bell
- Environmental Genomics Group, Genome Integrity and Structural Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
- Corresponding authors: . The Ludwig Institute for Cancer Research, The Nuffield Department of Clinical Medicine, The University of Oxford, Oxford, The United Kingdom. . Environmental Genomics Group, Genomic Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, MD C3-03, NIEHS, PO Box 12233, Research Triangle Park, NC 27709, The United States of America
| | - Gareth L. Bond
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
- Corresponding authors: . The Ludwig Institute for Cancer Research, The Nuffield Department of Clinical Medicine, The University of Oxford, Oxford, The United Kingdom. . Environmental Genomics Group, Genomic Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, MD C3-03, NIEHS, PO Box 12233, Research Triangle Park, NC 27709, The United States of America
| |
Collapse
|
196
|
Gusev A, Ko A, Shi H, Bhatia G, Chung W, Penninx BWJH, Jansen R, de Geus EJC, Boomsma DI, Wright FA, Sullivan PF, Nikkola E, Alvarez M, Civelek M, Lusis AJ, Lehtimäki T, Raitoharju E, Kähönen M, Seppälä I, Raitakari OT, Kuusisto J, Laakso M, Price AL, Pajukanta P, Pasaniuc B. Integrative approaches for large-scale transcriptome-wide association studies. Nat Genet 2016; 48:245-52. [PMID: 26854917 DOI: 10.1038/ng.3506] [Citation(s) in RCA: 1457] [Impact Index Per Article: 161.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 01/14/2016] [Indexed: 02/07/2023]
Abstract
Many genetic variants influence complex traits by modulating gene expression, thus altering the abundance of one or multiple proteins. Here we introduce a powerful strategy that integrates gene expression measurements with summary association statistics from large-scale genome-wide association studies (GWAS) to identify genes whose cis-regulated expression is associated with complex traits. We leverage expression imputation from genetic data to perform a transcriptome-wide association study (TWAS) to identify significant expression-trait associations. We applied our approaches to expression data from blood and adipose tissue measured in ∼ 3,000 individuals overall. We imputed gene expression into GWAS data from over 900,000 phenotype measurements to identify 69 new genes significantly associated with obesity-related traits (BMI, lipids and height). Many of these genes are associated with relevant phenotypes in the Hybrid Mouse Diversity Panel. Our results showcase the power of integrating genotype, gene expression and phenotype to gain insights into the genetic basis of complex traits.
Collapse
Affiliation(s)
- Alexander Gusev
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Arthur Ko
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Huwenbo Shi
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, California, USA
| | - Gaurav Bhatia
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Wonil Chung
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Brenda W J H Penninx
- Department of Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Rick Jansen
- Department of Psychiatry, VU University Medical Center, Amsterdam, the Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, VU University, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University, Amsterdam, the Netherlands
| | - Fred A Wright
- Bioinformatics Research Center, Department of Statistics, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Patrick F Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elina Nikkola
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Mete Civelek
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Pirkanmaa Hospital District and University of Tampere School of Medicine, Tampere, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Bogdan Pasaniuc
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, California, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
197
|
A Burden of Rare Variants Associated with Extremes of Gene Expression in Human Peripheral Blood. Am J Hum Genet 2016; 98:299-309. [PMID: 26849112 DOI: 10.1016/j.ajhg.2015.12.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/30/2015] [Indexed: 11/20/2022] Open
Abstract
In order to evaluate whether rare regulatory variants in the vicinity of promoters are likely to impact gene expression, we conducted a novel burden test for enrichment of rare variants at the extremes of expression. After sequencing 2-kb promoter regions of 472 genes in 410 healthy adults, we performed a quadratic regression of rare variant count on bins of peripheral blood transcript abundance from microarrays, summing over ranks of all genes. After adjusting for common eQTLs and the major axes of gene expression covariance, a highly significant excess of variants with minor allele frequency less than 0.05 at both high and low extremes across individuals was observed. Further enrichment was seen in sites annotated as potentially regulatory by RegulomeDB, but a deficit of effects was associated with known metabolic disease genes. The main result replicates in an independent sample of 75 individuals with RNA-seq and whole-genome sequence information. Three of four predicted large-effect sites were validated by CRISPR/Cas9 knockdown in K562 cells, but simulations indicate that effect sizes need not be unusually large to produce the observed burden. Unusually divergent low-frequency promoter haplotypes were observed at 31 loci, at least 9 of which appear to be derived from Neandertal admixture, but these were not associated with divergent gene expression in blood. The overall burden test results are consistent with rare and private regulatory variants driving high or low transcription at specific loci, potentially contributing to disease.
Collapse
|
198
|
Kuosmanen SM, Viitala S, Laitinen T, Peräkylä M, Pölönen P, Kansanen E, Leinonen H, Raju S, Wienecke-Baldacchino A, Närvänen A, Poso A, Heinäniemi M, Heikkinen S, Levonen AL. The Effects of Sequence Variation on Genome-wide NRF2 Binding--New Target Genes and Regulatory SNPs. Nucleic Acids Res 2016; 44:1760-75. [PMID: 26826707 PMCID: PMC4770247 DOI: 10.1093/nar/gkw052] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 01/16/2016] [Indexed: 12/11/2022] Open
Abstract
Transcription factor binding specificity is crucial for proper target gene regulation. Motif discovery algorithms identify the main features of the binding patterns, but the accuracy on the lower affinity sites is often poor. Nuclear factor E2-related factor 2 (NRF2) is a ubiquitous redox-activated transcription factor having a key protective role against endogenous and exogenous oxidant and electrophile stress. Herein, we decipher the effects of sequence variation on the DNA binding sequence of NRF2, in order to identify both genome-wide binding sites for NRF2 and disease-associated regulatory SNPs (rSNPs) with drastic effects on NRF2 binding. Interactions between NRF2 and DNA were studied using molecular modelling, and NRF2 chromatin immunoprecipitation-sequence datasets together with protein binding microarray measurements were utilized to study binding sequence variation in detail. The binding model thus generated was used to identify genome-wide binding sites for NRF2, and genomic binding sites with rSNPs that have strong effects on NRF2 binding and reside on active regulatory elements in human cells. As a proof of concept, miR-126–3p and -5p were identified as NRF2 target microRNAs, and a rSNP (rs113067944) residing on NRF2 target gene (Ferritin, light polypeptide, FTL) promoter was experimentally verified to decrease NRF2 binding and result in decreased transcriptional activity.
Collapse
Affiliation(s)
- Suvi M Kuosmanen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Sari Viitala
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Tuomo Laitinen
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Mikael Peräkylä
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Petri Pölönen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Emilia Kansanen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Hanna Leinonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Suresh Raju
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | | | - Ale Närvänen
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Antti Poso
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Anna-Liisa Levonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| |
Collapse
|
199
|
Watson CT, Roussos P, Garg P, Ho DJ, Azam N, Katsel PL, Haroutunian V, Sharp AJ. Genome-wide DNA methylation profiling in the superior temporal gyrus reveals epigenetic signatures associated with Alzheimer's disease. Genome Med 2016; 8:5. [PMID: 26803900 PMCID: PMC4719699 DOI: 10.1186/s13073-015-0258-8] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 12/29/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Alzheimer's disease affects ~13% of people in the United States 65 years and older, making it the most common neurodegenerative disorder. Recent work has identified roles for environmental, genetic, and epigenetic factors in Alzheimer's disease risk. METHODS We performed a genome-wide screen of DNA methylation using the Illumina Infinium HumanMethylation450 platform on bulk tissue samples from the superior temporal gyrus of patients with Alzheimer's disease and non-demented controls. We paired a sliding window approach with multivariate linear regression to characterize Alzheimer's disease-associated differentially methylated regions (DMRs). RESULTS We identified 479 DMRs exhibiting a strong bias for hypermethylated changes, a subset of which were independently associated with aging. DMR intervals overlapped 475 RefSeq genes enriched for gene ontology categories with relevant roles in neuron function and development, as well as cellular metabolism, and included genes reported in Alzheimer's disease genome-wide and epigenome-wide association studies. DMRs were enriched for brain-specific histone signatures and for binding motifs of transcription factors with roles in the brain and Alzheimer's disease pathology. Notably, hypermethylated DMRs preferentially overlapped poised promoter regions, marked by H3K27me3 and H3K4me3, previously shown to co-localize with aging-associated hypermethylation. Finally, the integration of DMR-associated single nucleotide polymorphisms with Alzheimer's disease genome-wide association study risk loci and brain expression quantitative trait loci highlights multiple potential DMRs of interest for further functional analysis. CONCLUSION We have characterized changes in DNA methylation in the superior temporal gyrus of patients with Alzheimer's disease, highlighting novel loci that facilitate better characterization of pathways and mechanisms underlying Alzheimer's disease pathogenesis, and improve our understanding of epigenetic signatures that may contribute to the development of disease.
Collapse
Affiliation(s)
- Corey T Watson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 3), James J. Peters VA Medical Center, Bronx, NY, USA
| | - Paras Garg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel J Ho
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nidha Azam
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pavel L Katsel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 3), James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew J Sharp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
200
|
|