Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Multi-ethnic genome-wide association study for atrial fibrillation

Subjects

This article has been updated

Abstract

Atrial fibrillation (AF) affects more than 33 million individuals worldwide1 and has a complex heritability2. We conducted the largest meta-analysis of genome-wide association studies (GWAS) for AF to date, consisting of more than half a million individuals, including 65,446 with AF. In total, we identified 97 loci significantly associated with AF, including 67 that were novel in a combined-ancestry analysis, and 3 that were novel in a European-specific analysis. We sought to identify AF-associated genes at the GWAS loci by performing RNA-sequencing and expression quantitative trait locus analyses in 101 left atrial samples, the most relevant tissue for AF. We also performed transcriptome-wide analyses that identified 57 AF-associated genes, 42 of which overlap with GWAS loci. The identified loci implicate genes enriched within cardiac developmental, electrophysiological, contractile and structural pathways. These results extend our understanding of the biological pathways underlying AF and may facilitate the development of therapeutics for AF.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Study and analysis flowchart.
Fig. 2: Manhattan plot of combined-ancestry meta-analysis.
Fig. 3: Volcano plot of transcriptome-wide analysis from human heart tissues.
Fig. 4: Cross-trait associations of AF risk variants with AF risk factors in the UK Biobank.

Similar content being viewed by others

Change history

  • 20 July 2018

    In the version of this article initially published, Supplementary Tables 1, 2, 6, 8, 10 and 19–22 and the Supplementary Note were omitted from the supplementary PDF. The supplementary PDF now includes these items.

References

  1. Chugh, S. S. et al. Worldwide epidemiology of atrial fibrillation: a Global Burden of Disease 2010 study. Circulation 129, 837–847 (2014).

    PubMed  Google Scholar 

  2. Lubitz, S. A. et al. Association between familial atrial fibrillation and risk of new-onset atrial fibrillation. JAMA 304, 2263–2269 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. January, C. T. et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: executive summary. J. Am. Coll. Cardiol. 64, 2071–2104 (2014).

    Google Scholar 

  4. Benjamin, E. J. et al. Variants in ZFHX3 are associated with atrial fibrillation in individuals of European ancestry. Nat. Genet. 41, 879–881 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Ellinor, P. T. et al. Meta-analysis identifies six new susceptibility loci for atrial fibrillation. Nat. Genet. 44, 670–675 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Sinner, M. F. et al. Integrating genetic, transcriptional, and functional analyses to identify 5 novel genes for atrial fibrillation. Circulation 130, 1225–1235 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ellinor, P. T. et al. Common variants in KCNN3 are associated with lone atrial fibrillation. Nat. Genet. 42, 240–244 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Christophersen, I. E. et al. Large-scale analyses of common and rare variants identify 12 new loci associated with atrial fibrillation. Nat. Genet. 49, 946–952 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Low, S.-K. et al. Identification of six new genetic loci associated with atrial fibrillation in the Japanese population. Nat. Genet. 49, 953–958 (2017).

    CAS  PubMed  Google Scholar 

  10. Weng, L.-C. et al. Heritability of atrial fibrillation. Circ. Cardiovasc. Genet. 10, e001838 (2017).

    PubMed  PubMed Central  Google Scholar 

  11. Klarin, D. et al. Genetic analysis in UK Biobank links insulin resistance and transendothelial migration pathways to coronary artery disease. Nat. Genet. 49, 1392–1397 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Barbeira, A. N. et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat. Commun. 9, 1825 (2018).

    PubMed  PubMed Central  Google Scholar 

  13. Sudlow, C. et al. UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 12, e1001779 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Lu, X. et al. Genome-wide association study in Chinese identifies novel loci for blood pressure and hypertension. Hum. Mol. Genet. 24, 865–74 (2015).

    CAS  PubMed  Google Scholar 

  15. Nielsen, J. B. et al. Genome-wide association study of 1 million people identifies 111 loci for atrial fibrillation. Preprint at https://www.biorxiv.org/content/early/2018/01/04/242149 (2018).

  16. Sinner, M. F. et al. The non-synonymous coding IKr-channel variant KCNH2-K897T is associated with atrial fibrillation: results from a systematic candidate gene-based analysis of KCNH2 (HERG). Eur. Heart J. 29, 907–914 (2008).

    CAS  PubMed  Google Scholar 

  17. Olson, T. M. et al. Sodium channel mutations and susceptibility to heart failure and atrial fibrillation. JAMA 293, 447–454 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. McNair, W. P. et al. SCN5A mutation associated with dilated cardiomyopathy, conduction disorder, and arrhythmia. Circulation 110, 2163–2167 (2004).

    CAS  PubMed  Google Scholar 

  19. van Weerd, J. H. et al. A large permissive regulatory domain exclusively controls Tbx3 expression in the cardiac conduction system. Circ. Res. 115, 432–441 (2014).

    PubMed  Google Scholar 

  20. Schott, J. J. et al. Congenital heart disease caused by mutations in the transcription factor NKX2-5. Science 281, 108–101 (1998).

    CAS  PubMed  Google Scholar 

  21. den Hoed, M. et al. Identification of heart rate–associated loci and their effects on cardiac conduction and rhythm disorders. Nat. Genet. 45, 621–631 (2013).

    Google Scholar 

  22. Kirchhof, P. et al. PITX2c is expressed in the adult left atrium, and reducing Pitx2c expression promotes atrial fibrillation inducibility and complex changes in gene expression. Circ. Cardiovasc. Genet. 4, 123–133 (2011).

    CAS  PubMed  Google Scholar 

  23. Wang, J. et al. Pitx2 prevents susceptibility to atrial arrhythmias by inhibiting left-sided pacemaker specification. Proc. Natl. Acad. Sci. USA 107, 9753–9758 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Syeda, F. et al. PITX2 modulates atrial membrane potential and the antiarrhythmic effects of sodium-channel blockers. J. Am. Coll. Cardiol. 68, 1881–1894 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Nadadur, R. D. et al. Pitx2 modulates a Tbx5-dependent gene regulatory network to maintain atrial rhythm. Sci. Transl. Med. 8, 354ra115 (2016).

    PubMed  PubMed Central  Google Scholar 

  26. Tucker, N. R. et al. Diminished PRRX1 expression is associated with increased risk of atrial fibrillation and shortening of the cardiac action potential. Circ. Cardiovasc. Genet. 10, e001902 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Postma, A. V. et al. A gain-of-function TBX5 mutation is associated with atypical Holt–Oram syndrome and paroxysmal atrial fibrillation. Circ. Res. 102, 1433–1442 (2008).

    CAS  PubMed  Google Scholar 

  28. Lahat, H. et al. A missense mutation in a highly conserved region of CASQ2 is associated with autosomal recessive catecholamine-induced polymorphic ventricular tachycardia in Bedouin families from Israel. Am. J. Hum. Genet. 69, 1378–1384 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Lahat, H. et al. Autosomal recessive catecholamine- or exercise-induced polymorphic ventricular tachycardia: clinical features and assignment of the disease gene to chromosome 1p13-21. Circulation 103, 2822–2827 (2001).

    CAS  PubMed  Google Scholar 

  30. Corrado, D., Link, M. S. & Calkins, H. Arrhythmogenic right ventricular cardiomyopathy. N. Engl. J. Med. 376, 61–72 (2017).

    CAS  PubMed  Google Scholar 

  31. Gerull, B. et al. Mutations in the desmosomal protein plakophilin-2 are common in arrhythmogenic right ventricular cardiomyopathy. Nat. Genet. 36, 1162–1164 (2004).

    CAS  PubMed  Google Scholar 

  32. Ackerman, M. J. et al. HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies. Heart Rhythm 8, 1308–1339 (2011).

    PubMed  Google Scholar 

  33. Weng, L.-C. et al. Genetic predisposition, clinical risk factor burden, and lifetime risk of atrial fibrillation. Circulation 137, 1027–1038 (2017).

    PubMed  PubMed Central  Google Scholar 

  34. Korn, J. M. et al. Integrated genotype calling and association analysis of SNPs, common copy number polymorphisms and rare CNVs. Nat. Genet. 40, 1253–1260 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Goldstein, J. I. et al. zCall: a rare variant caller for array-based genotyping. Bioinformatics 28, 2543–2545 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Pulit, S. L. et al. Loci associated with ischaemic stroke and its subtypes (SiGN): a genome-wide association study. Lancet Neurol. 15, 174–184 (2016).

    Google Scholar 

  37. Bycroft, C. et al. Genome-wide genetic data on ~500,000 UK Biobank participants. Preprint at https://www.biorxiv.org/content/early/2017/07/20/166298 (2017).

  38. The Haplotype Reference Consortium et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 48, 1279–1283 (2016).

    Google Scholar 

  39. Das, S. et al. Next-generation genotype imputation service and methods. Nat. Genet. 48, 1284–1287 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Auton, A. et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    PubMed  Google Scholar 

  41. Francioli, L. C. et al. Whole-genome sequence variation, population structure and demographic history of the Dutch population. Nat. Genet. 46, 818–825 (2014).

    CAS  Google Scholar 

  42. Chang, C. C. et al. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 4, 7 (2015).

    PubMed  PubMed Central  Google Scholar 

  43. Alexander, D. H., Novembre, J. & Lange, K. Fast model-based estimation of ancestry in unrelated individuals. Genome Res. 19, 1655–1664 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Price, A. L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    CAS  PubMed  Google Scholar 

  45. Bellenguez, C., Strange, A., Freeman, C., Donnelly, P. & Spencer, C. C. A. A robust clustering algorithm for identifying problematic samples in genome-wide association studies. Bioinformatics 28, 134–135 (2012).

    CAS  PubMed  Google Scholar 

  46. Aulchenko, Y. S., Struchalin, M. V. & van Duijn, C. M. ProbABEL package for genome-wide association analysis of imputed data. BMC Bioinformatics 11, 134 (2010).

    PubMed  PubMed Central  Google Scholar 

  47. Marchini, J., Howie, B., Myers, S., McVean, G. & Donnelly, P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat. Genet. 39, 906–913 (2007).

    CAS  PubMed  Google Scholar 

  48. Chanda, P., Huang, H., Arking, D. E. & Bader, J. S. Fast association tests for genes with FAST. PLoS One 8, e68585 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. R Core Team. R: A Language and Environment for Statistical Computing. http://www.r-project.org/ (2015).

  50. Willer, C. J., Li, Y. & Abecasis, G. R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Fadista, J., Manning, A. K., Florez, J. C. & Groop, L. The (in)famous GWAS P-value threshold revisited and updated for low-frequency variants. Eur. J. Hum. Genet. 24, 1202–1205 (2016).

    PubMed  PubMed Central  Google Scholar 

  52. Higgins, J. P. T., Thompson, S. G., Deeks, J. J. & Altman, D. G. Measuring inconsistency in meta-analyses. Br. Med. J. 327, 557–560 (2003).

    Google Scholar 

  53. McLaren, W. et al. The Ensembl Variant Effect Predictor. Genome Biol. 17, 122 (2016).

    PubMed  PubMed Central  Google Scholar 

  54. Schwarz, J. M., Rödelsperger, C., Schuelke, M. & Seelow, D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat. Methods 7, 575–576 (2010).

    CAS  PubMed  Google Scholar 

  55. Kumar, P., Henikoff, S. & Ng, P. C. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 4, 1073–1081 (2009).

    CAS  PubMed  Google Scholar 

  56. Chun, S. & Fay, J. C. Identification of deleterious mutations within three human genomes. Genome Res. 19, 1553–1561 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Adzhubei, I. A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ernst, J. & Kellis, M. Large-scale imputation of epigenomic datasets for systematic annotation of diverse human tissues. Nat. Biotechnol. 33, 364–376 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Ward, L. D. & Kellis, M. HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res. 40, D930–D934 (2012).

    CAS  PubMed  Google Scholar 

  60. Pers, T. H., Timshel, P. & Hirschhorn, J. N. SNPsnap: a Web-based tool for identification and annotation of matched SNPs. Bioinformatics 31, 418–420 (2015).

    CAS  PubMed  Google Scholar 

  61. Fay, M. P. & Shaw, P. A. Exact and asymptotic weighted logrank tests for interval censored data: the interval R package. J. Stat. Softw. 36, 1–34 (2010).

    Google Scholar 

  62. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  63. Harrow, J. et al. GENCODE: The reference human genome annotation for The ENCODE Project. Genome Res. 22, 1760–1774 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Delaneau, O. et al. A complete tool set for molecular QTL discovery and analysis. Nat. Commun. 8, 15452 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. GTEx Consortium. The Genotype–Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

  66. Aguet, F. et al. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    Google Scholar 

  67. Gamazon, E. R. et al. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet. 47, 1091–1098 (2015).

  68. Yang, J. et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 44, 369–375 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Yang, J., Lee, S. H., Goddard, M. E. & Visscher, P. M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Segrè, A. V. et al. Common inherited variation in mitochondrial genes is not enriched for associations with type 2 diabetes or related glycemic traits. PLoS Genet. 6, e1001058 (2010).

    PubMed  PubMed Central  Google Scholar 

  71. Welter, D. et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res. 42, D1001–D1006 (2014).

    CAS  PubMed  Google Scholar 

  72. MacArthur, J. et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 45, D896–D901 (2017).

    CAS  PubMed  Google Scholar 

  73. Loh, P.-R. et al. Contrasting genetic architectures of schizophrenia and other complex diseases using fast variance-components analysis. Nat. Genet. 47, 1385–1392 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

A full list of acknowledgments appears in the Supplementary Notes.

Author information

Authors and Affiliations

Authors

Contributions

C.R., M.D.C., E.J.B., K.L.L., S.A.L., P.T.E. and H.L. drafted and finalized the manuscript. H.J.C., E.A.D., B.L.K., B. Weijs, S. Kääb, M.M.-N., B.N., K.S., M.F.S., J.L., A.A., L.Y.C., K.L., S.A., D.C., G.P., L. Risch, S. Thériault, T.T., C. Schurman, S.A.S., J.C.D., D.M.R., Q.S.W., C.R., M.D.C., K.G.A., B.R.D., N.G., S. Kathiresan, L.M., P.L.H., J.B., M.K.C., J.D.S., H. Sun, D.R.V.W., T.M.B., J.C.B., J.A.B., G.A., M.S.O., L. Refsgaard, J.H.S., D.F., R.J., S. Shah, P.K., R.B.S., T.E., M.T.-L., E.J.B., B. Wang, K.L.L., M. Kähönen, T.L., I.E.C., I.C.V.G., B.G., M. Rienstra, J.E.S., P.V.D.H., N.V., H.L.B., S.C.D., R.G., B.L., S. Saba, A.A.S., R.W., H.C., R.N.L., N.L.S., K.L.W., S.R.H., B.M.P., N.S., J. Carlquist, M.J.C., S. Knight, M.E.K., W.M., P.A., O.M., M.O.-M., X.G., H.J.L., J.I.R., K.D.T., S.H.C., N.R.T., S.A.L., P.T.E., C.N.-C., M.A.R., C.D.A., P.N., J.J.G.S., H. Schunkert, T.P.C., K.B.M., I.F., J.J.W.J., P.W.M., R.N., S. Trompet, O.H.F., A. Hofman, M. Kavousi, M.N.N., B.H. Stricker, A.G.U., R.P.G., J.J.-C., S.L.P., S.M., A. Hamsten, J.P.K., G.M.M., C.R.P., A.P.M., S.G., E. Ingelsson, H.L., D.D., J.A.M., M.M.B.S., Z.T.Y., C. Shaffer, P.E.W., C.M.A., D.I.C., R.K.S., J.W., M. Dichgans and R.M. contributed to and revised the manuscript. H.J.C., E.A.D., B.L.K., B. Weijs, S. Kääb, M.M.-N., B.N., K.S., M.F.S., V.G., T.B.H., L.J.L., A.V.S., M.E., J. Hernesniemi, J.L., I.S., A.A., D.E.A., N.A.B., E.B., L.Y.C., M.L., E.Z.S., S.A., D.C., G.P., L. Risch, S. Thériault, K.I., Y.K., M. Kubo, S.-K.L., T.T., E.B.B., R.J.F.L., Y.L., C. Schurman, S.A.S., J.C.D., D.M.R., Q.S.W., C.R., M.D.C., L.-C.W., K.G.A., N.G., S. Kathiresan, L.M., P.L.H., J.B., M.K.C., J.D.S., H. Sun, D.R.V.W., T.M.B., J.C.B., J.A.B., M.-L.L., J. Sinisalo, E.V., G.A., M.S.O., L. Refsgaard, J.H.S., D.F., R.J., A. Sun, P.K., H.O., R.B.S., T.Z., T.E., M.T.-L., E.J.B., B. Wang, K.L.L., M. Kähönen, T.L., L.-P.L., K.N., I.E.C., A. Tveit, B.G., J.E.S., N.V., H.L.B., S.C.D., R.G., B.L., S. Saba, A.A.S., R.W., A.C., C.H., L.J.H., J. Huffman, S.P., D.P., B.H. Smith, H.C., E. Ipek, S.N., R.N.L., N.L.S., K.L.W., S.R.H., B.M.P., N.S., J. Carlquist, M.J.C., S. Knight, E.-K.C., H.E.L., H.-N.P., J. Shim, P.-S.Y., G.D., J. Huang, M.E.K., P.A., O.M., M.O.-M., Y.-D.C., X.G., K.D.T., J.Y., S.A.L., P.T.E., C.N.-C., M.A.R., J.R., N.R., C.D.A., P.N., J.J.G.S., A.K., T.K., H. Schunkert, L.Z., T.P.C., S.M.D., K.B.M., M.P.M., D.J.R., I.F., J.J.W.J., S. Trompet, O.H.F., A. Hofman, M. Kavousi, M.N.N., B.H. Stricker, A.G.U., M. Dörr, S.B.F., A. Teumer, U.V., S.W., J.W.C., R.P.G., J.J.-C., P.K.-W., J.P., S.L.P., M. Ribasés, A. Slowik, D.W., B.B.W., A.R.V.R.H., J.E.K., A.J.M., A.P., S.M., A.N., A. Hamsten, P.K.M., N.L.P., J.P.K., G.M.M., C.R.P., J. Cook, L.L., C.M.L., A.M., A.P.M., S.G., E. Ingelsson, N.E., K.T., H.L., D.D.M., D.D., J.A.M., M.M.B.S., Z.T.Y., C. Shaffer, P.E.W., C.M.A., D.I.C., P.M.R., M. Dichgans and R.M. contributed to study-specific GWAS by providing phenotype data or performing data analyses. C.R., M.D.C. and S.L.P. performed meta-analyses. N.R.T., P.T.E., T.P.C., K.B.M., M.P.M. and H.L. contributed samples sequencing or performed left atrial eQTL analyses. C.R., M.D.C., L.-C.W., K.L.L., S.H.C., N.R.T. and H.L. performed downstream analyses. K.I., T.T., K.L.L., S.R.H., S.A.L. and P.T.E. conceived designed and supervised the overall project.

Corresponding author

Correspondence to Patrick T. Ellinor.

Ethics declarations

Competing interests

P.T.E is the PI on a grant from Bayer to the Broad Institute focused on the genetics and therapeutics of AF. B.M.P. serves on the DSMB of a clinical trial funded by Zoll LifeCor and on the Steering Committee of the Yale Open Data Access Project funded by Johnson & Johnson. P.K. receives research support from the European Union, the British Heart Foundation, the Leducq Foundation, the Medical Research Council (UK) and the German Centre for Cardiovascular Research, and from several drug and device companies active in AF, and has received honoraria from several such companies. P.K. is also listed as an inventor on two patents held by University of Birmingham (Atrial Fibrillation Therapy WO 2015140571, Markers for Atrial Fibrillation WO 2016012783). K.L. is an employee of Bayer. The genotyping of participants in the Broad AF study and the expression analysis of LA tissue samples were supported by a grant from Bayer to the Broad Institute. S.N. is a consultant to Biosense Webster, Siemens and Cardiosolv. S.N. also receives research grants from NIH/NHLBI, Siemens, Biosense Webster and Imricor. S. Kathiresan has received grant support from Bayer and Amarin; holds equity in San Therapeutics and Catabasis; and has received personal fees for participation in scientific advisory boards for Catabasis, Regeneron Genetics Center, Merck, Celera, Genomics PLC, Corvidia Therapeutics and Novo Ventures. S. Kathiresan also received personal fees for consulting services from Novartis, AstraZeneca, Alnylam, Eli Lilly Company, Leerink Partners, Merck, Noble Insights, Bayer, Ionis Pharmaceuticals, Novo Ventures, Haug Partners LLC and Genetic Modifiers Newco, Inc. S.A.L. receives sponsored research support from Bristol Myers Squibb, Bayer, Biotronik and Boehringer Ingelheim, and has consulted for St. Jude Medical/Abbott and Quest Diagnostics. The remaining authors have no disclosures.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated Supplementary Information

Supplementary Figure 1 Quantile–quantile plot of combined ancestry meta-analysis.

Quantile–quantile plot of combined ancestry meta-analysis for n = 12,149,979 included variants and λGC = 1.0948.

Supplementary Figure 2 Venn diagram for genes near sentinel variants from combined ancestry meta-analysis within enriched gene sets, by functional groups.

The Venn diagram shows genes that are within enriched gene set from the gene set enrichment analysis and within 500 kb of a sentinel variant. The genes were manually grouped into functional categories based on their corresponding gene sets. The diagram shows the overlap between the genes and the functional categories.

Supplementary Figure 3 Manhattan plot of European-ancestry meta-analysis.

The plot shows novel (red and purple) and known (blue) genetic loci associated with AF at a significance level of P < 1 × 10–8 (dotted line) for the European-ancestry meta-analysis (n = 537,409). The significance level accounts for multiple testing of independent variants with MAF ≥0.1% using a Bonferroni correction. P values (two-sided) were derived from a meta-analysis using a fixed-effects model with an inverse-variance-weighted approach. Loci in purple did not reach genome-wide significance in the combined ancestry meta-analysis. Gene labels correspond to the nearest gene(s). The y axis has a break between –log10 (P) of 25 and 400 to emphasize the novel loci.

Supplementary Figure 4 Quantile–quantile plot of European-ancestry meta-analysis.

Quantile–quantile plot of European-ancestry meta-analysis for n = 9,362,422 included variants and λGC = 1.1194.

Supplementary Figure 5 Manhattan plot of African-American meta-analysis.

The plot shows known (blue) genetic loci associated with AF at a significance level of P < 1 × 10–8 (dotted line), for the African-American-ancestry meta-analysis (n = 8,967). The significance level accounts for multiple testing of independent variants with MAF ≥ 0.1% using a Bonferroni correction. P values (two-sided) were derived from a meta-analysis using a fixed-effects model with an inverse-variance-weighted approach. The gene label corresponds to the nearest gene.

Supplementary Figure 6 Quantile–quantile plot of African-American-ancestry meta-analysis.

Quantile–quantile plot of African-American-ancestry meta-analysis for n = 8,640,046 included variants and λGC = 0.997.

Supplementary Figure 7 Regional plots for 4q25 for European, Japanese and African American ancestry and pairwise LD between sentinel variants at 4q25.

ac, Regional plots of 4q25 for European-ancestry results (a, n = 537,409), Japanese-ancestry results (b, n = 36,792) and African-American-ancestry results (c, n = 8,967). LD is shown based on the 1000 Genomes phase 1 v3 reference, using the populations EUR (a), ASN (b) and AFR (c). d, Pairwise LD (r2) for the sentinel variants based on the LD from the 1000 Genomes phase 1 v3 reference for EUR (n = 379), ASN (n = 286) and AFR (n = 246) ancestry. 1000G, 1000 Genomes; AA, African American; AFR, African; ASN, Asian; EUR, European; JAP, Japanese; LD, linkage disequilibrium.

Supplementary Figure 8 Forest plots of odds ratios, and allele frequency plots, by ancestry for sentinel variants with significant heterogeneity.

ac, Forest plots of odds ratios and pie charts of allele frequencies across ancestries (EUR, n = 537,409; JAP, n = 36,792; AA, n = 8,967; BRAZ, n = 1,664; HISP, n = 3,358) for sentinel variants with significant heterogeneity, close to PITX2 (a), NEURL (b) and ZFHX3 (c). Shown are odds ratios with 95% confidence intervals. Frequencies of the effect allele are depicted in blue; frequencies of the reference allele are depicted in orange. AA, African American; BRAZ, Brazilian; EUR, European; HISP, Hispanic; JAP, Japanese.

Supplementary Figure 9 Enrichment of atrial fibrillation–associated loci across ChromHMM regulatory regions.

a,b, Percent overlap of loci with regulatory regions (promoter, enhancer, DNase) based on the Roadmap Epigenomics Consortium 25-state model across all tissues (a) and cardiac tissues (b). Each locus includes sentinel variant and proxies with r2 > 0.6. The P values were derived from one-tailed permutation tests (n = 1,000). 1000 Genomes control loci were matched to atrial fibrillation sentinel SNPs via SNPSnap (n = 93,000). Atrial fibrillation–associated loci are from the combined ancestry analysis (n = 93). The sentinel SNP for one AF locus could not be matched in SNPSnap and was excluded from this analysis. The box plot depicts the following values: the center represents the median, the top and bottom of the box represent the first and third quartile, the whiskers reach to 1.5 times the interquartile range, and data points outside the whiskers are plotted as outliers. *P = 0.001. 1000G, 1000 Genomes; AF, atrial fibrillation.

Supplementary information

Supplementary Text, Figures and Tables

Supplementary Figures 1–9, Supplementary Tables 1, 2, 6, 8, 10 and 19–22, and Supplementary Notes

Reporting Summary

Supplementary Table 3

Known loci in combined ancestry meta-analysis

Supplementary Table 4

Gene set enrichment analysis results for combined ancestry meta-analysis

Supplementary Table 5

Novel and known loci in ancestry-specific meta-analyses

Supplementary Table 7

Loci with multiple signals identified by conditional and joint analysis for European-ancestry meta-analysis

Supplementary Table 9

Chromatin states for sentinel variants and proxies from Roadmap Epigenomics across all tissues and heart

Supplementary Table 11

Significant cis-eQTLs for sentinel variants from combined ancestry meta-analysis in GTEx heart tissues

Supplementary Table 12

Probable AF susceptibility genes for loci from combined ancestry meta-analysis

Supplementary Table 13

Transcriptome-wide results based on summary-level data from combined ancestry meta-analysis

Supplementary Table 14

Association to diseases and traits in NHGRI-EBI GWAS catalog for sentinel variants or proxies from combined ancestry meta-analysis

Supplementary Table 15

PheWAS results in UK Biobank for sentinel variants from combined ancestry meta-analysis

Supplementary Table 16

134 loci associated with atrial fibrillation

Supplementary Table 17

Baseline summary for GWAS

Supplementary Table 18

GWAS summary on genotyping, QC, imputation and analysis per study

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roselli, C., Chaffin, M.D., Weng, LC. et al. Multi-ethnic genome-wide association study for atrial fibrillation. Nat Genet 50, 1225–1233 (2018). https://doi.org/10.1038/s41588-018-0133-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0133-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing