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:

Meta-analysis of genome-wide associations and polygenic risk prediction for atrial fibrillation in more than 180,000 cases

Abstract

Atrial fibrillation (AF) is the most common heart rhythm abnormality and is a leading cause of heart failure and stroke. This large-scale meta-analysis of genome-wide association studies increased the power to detect single-nucleotide variant associations and found more than 350 AF-associated genetic loci. We identified candidate genes related to muscle contractility, cardiac muscle development and cell–cell communication at 139 loci. Furthermore, we assayed chromatin accessibility using assay for transposase-accessible chromatin with sequencing and histone H3 lysine 4 trimethylation in stem cell-derived atrial cardiomyocytes. We observed a marked increase in chromatin accessibility for our sentinel variants and prioritized genes in atrial cardiomyocytes. Finally, a polygenic risk score (PRS) based on our updated effect estimates improved AF risk prediction compared to the CHARGE-AF clinical risk score and a previously reported PRS for AF. The doubling of known risk loci will facilitate a greater understanding of the pathways underlying 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: Miami plot of meta-analysis across 181,446 AF cases and 1,468,899 controls for common variants and low-frequency variants.
Fig. 2: Prioritization of genes at common variant AF GWAS loci.
Fig. 3: Polygenic risk prediction of AF with PRS in HUNT and UK Biobank.
Fig. 4: Phenome-wide associations of diseases and traits to the PRSAF in the UK Biobank.
Fig. 5: Phenome-wide associations of traits to the PRSAF in the UK Biobank.

Similar content being viewed by others

Data availability

The summary-level results file as well as the weights file for the PRSAF are available for download at the Cardiovascular Disease Knowledge Portal under the weblinks https://cvd.hugeamp.org/downloads.html#polygenic and https://cvd.hugeamp.org/downloads.html#summary. The raw and processed ATAC–seq and H3K4me3 data have been deposited at the NCBI Gene Expression Omnibus under accession number GSE225293. The following datasets were used in this study and are publicly available under the listed weblinks: GENCODE: https://www.gencodegenes.org; 1000G LD reference, MAGMA gene annotations and precomputed files for PoPS algorithm: https://www.dropbox.com/sh/o6t5jprvxb8b500/AADZ8qD6Rpz4uvCk0b5nUnPaa/data?dl=0; GTEx: https://www.gtexportal.org/home; ENCODE: https://www.encodeproject.org; OpenTargets: https://www.opentargets.org.

Code availability

All software programs used in the study are publicly available and described in the Methods and Reporting Summary.

References

  1. Chen, Y.-H. et al. KCNQ1 gain-of-function mutation in familial atrial fibrillation. Science 299, 251–254 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Hodgson-Zingman, D. M. et al. Atrial natriuretic peptide frameshift mutation in familial atrial fibrillation. N. Engl. J. Med. 359, 158–165 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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).

    Article  CAS  PubMed  Google Scholar 

  4. Orr, N. et al. A mutation in the atrial-specific myosin light chain gene (MYL4) causes familial atrial fibrillation. Nat. Commun. 7, 11303 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gudbjartsson, D. F. et al. Variants conferring risk of atrial fibrillation on chromosome 4q25. Nature 448, 353–357 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Thorolfsdottir, R. B. et al. Coding variants in RPL3L and MYZAP increase risk of atrial fibrillation. Commun. Biol. 1, 68 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nielsen, J. B. et al. Genome-wide study of atrial fibrillation identifies seven risk loci and highlights biological pathways and regulatory elements involved in cardiac development. Am. J. Hum. Genet. 102, 103–115 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Gudbjartsson, D. F. et al. A sequence variant in ZFHX3 on 16q22 associates with atrial fibrillation and ischemic stroke. Nat. Genet. 41, 876–878 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Roselli, C. et al. Multi-ethnic genome-wide association study for atrial fibrillation. Nat. Genet. 50, 1225–1233 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Nielsen, J. B. et al. Biobank-driven genomic discovery yields new insight into atrial fibrillation biology. Nat. Genet. 50, 1234–1239 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Sakaue, S. et al. A cross-population atlas of genetic associations for 220 human phenotypes. Nat. Genet. 53, 1415–1424 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Miyazawa, K. et al. Cross-ancestry genome-wide analysis of atrial fibrillation unveils disease biology and enables cardioembolic risk prediction. Nat. Genet. 55, 187–197 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. de Leeuw, C. A., Mooij, J. M., Heskes, T. & Posthuma, D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 11, e1004219 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Weeks, E. M. et al. Leveraging polygenic enrichments of gene features to predict genes underlying complex traits and diseases. Nat. Genet. 55, 1267–1276 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Choi, S. H. et al. Sequencing in over 50,000 cases identifies coding and structural variation underlying atrial fibrillation risk. Nat. Genet. https://doi.org/10.1038/s41588-025-02074-9 (2025).

  21. van Ouwerkerk, A. F. et al. Identification of atrial fibrillation associated genes and functional non-coding variants. Nat. Commun. 10, 4755 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Clausen, A. G., Vad, O. B., Andersen, J. H. & Olesen, M. S. Loss-of-function variants in the SYNPO2L gene are associated with atrial fibrillation. Front. Cardiovasc. Med. 8, 650667 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Choi, S. H. et al. Association between Titin loss-of-function variants and early-onset atrial fibrillation. JAMA 320, 2354 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Faggioni, M. et al. Suppression of spontaneous Ca elevations prevents atrial fibrillation in calsequestrin 2-null hearts. Circ. Arrhythm. Electrophysiol. 7, 313–320 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Purohit, A. et al. Oxidized Ca2+/calmodulin-dependent protein kinase II triggers atrial fibrillation. Circulation 128, 1748–1757 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Holm, H. et al. A rare variant in MYH6 is associated with high risk of sick sinus syndrome. Nat. Genet. 43, 316–320 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. McMullen, J. R. et al. The insulin-like growth factor 1 receptor induces physiological heart growth via the phosphoinositide 3-kinase(p110α) pathway. J. Biol. Chem. 279, 4782–4793 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. Wu, T. et al. HSPB7 is indispensable for heart development by modulating actin filament assembly. Proc. Natl Acad. Sci. USA 114, 11956–11961 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Khera, A. V. et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat. Genet. 50, 1219–1224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Vilhjálmsson, B. J. et al. Modeling linkage disequilibrium increases accuracy of polygenic risk scores. Am. J. Hum. Genet. 97, 576–592 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Alonso, A. et al. Simple risk model predicts incidence of atrial fibrillation in a racially and geographically diverse population: the CHARGE‐AF Consortium. J. Am. Heart Assoc. 2, e000102 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Ge, T., Chen, C.-Y., Ni, Y., Feng, Y.-C. A. & Smoller, J. W. Polygenic prediction via Bayesian regression and continuous shrinkage priors. Nat. Commun. 10, 1776 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Jurgens, S. J. et al. Analysis of rare genetic variation underlying cardiometabolic diseases and traits among 200,000 individuals in the UK Biobank. Nat. Genet. 54, 240–250 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Nagai, A. et al. Overview of the BioBank Japan Project: study design and profile. J. Epidemiol. 27, S2–S8 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Mountjoy, E. et al. An open approach to systematically prioritize causal variants and genes at all published human GWAS trait-associated loci. Nat. Genet. 53, 1527–1533 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bycroft, C. et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 562, 203–209 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kowalski, M. H. et al. Use of >100,000 NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium whole genome sequences improves imputation quality and detection of rare variant associations in admixed African and Hispanic/Latino populations. PLoS Genet. 15, e1008500 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Akiyama, M. et al. Genome-wide association study identifies 112 new loci for body mass index in the Japanese population. Nat. Genet. 49, 1458–1467 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  41. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Frankish, A. et al. GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res. 47, D766–D773 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Tucker, N. R. et al. Transcriptional and cellular diversity of the human heart. Circulation 142, 466–482 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Finucane, H. K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228–1235 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).

    Article  Google Scholar 

  46. Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Liu, X., Li, C., Mou, C., Dong, Y. & Tu, Y. dbNSFP v4: a comprehensive database of transcript-specific functional predictions and annotations for human nonsynonymous and splice-site SNVs. Genome Med. 12, 103 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ashburner, M. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kanehisa, M., Furumichi, M., Sato, Y., Ishiguro-Watanabe, M. & Tanabe, M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 49, D545–D551 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Jassal, B. et al. The reactome pathway knowledgebase. Nucleic Acids Res. 48, D498–D503 (2020).

    CAS  PubMed  Google Scholar 

  52. Köhler, S. et al. The Human Phenotype Ontology in 2021. Nucleic Acids Res. 49, D1207–D1217 (2021).

    Article  PubMed  Google Scholar 

  53. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  Google Scholar 

  54. Martens, M. et al. WikiPathways: connecting communities. Nucleic Acids Res. 49, D613–D621 (2021).

    Article  CAS  PubMed  Google Scholar 

  55. Corces, M. R. et al. An improved ATAC–seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kent, W. J. et al. The Human Genome Browser at UCSC. Genome Res. 12, 996–1006 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, e21856 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb.) 2, 100141 (2021).

    CAS  PubMed  Google Scholar 

  60. Krokstad, S. et al. Cohort profile: the HUNT study, Norway. Int. J. Epidemiol. 42, 968–977 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Staerk, L. et al. Lifetime risk of atrial fibrillation according to optimal, borderline, or elevated levels of risk factors: cohort study based on longitudinal data from the Framingham Heart Study. Br. Med. J. 361, k1453 (2018).

    Article  Google Scholar 

Download references

Acknowledgements

The GTEx Project was supported by the Common Fund of the Office of the Director of the National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH and NINDS. The data used for the analyses described in this manuscript were obtained from the GTEx Portal on 02/10/2021. This research is based on data from the MVP, Office of Research and Development, Veterans Health Administration and was supported by award no. I01-BX004821. This publication does not necessarily represent the views of the Department of Veteran Affairs or the United States Government. Study-specific acknowledgements, including grants, are listed in the Supplementary Note.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

C.R., C.W., D.G., G.S., H.H., I.S., K.S., M.C., M.C.H., M.S.O., N.A.M. and P.T.E. crafted and finalized the manuscript. A.A., A.C., A.C.P., A.N., A.T., B.B., B.G., B.H.S., B.M.P., B.S., C.D.A., C.E., C.G., C. Haggerty, C. Hayward, C.R., C.R.P., C.T.R., C.W., D.O.A., D.C., D.D.M., D.E.A., D.F., D.F.F., D.G., D.I.C., D.J.R., D.L., D.S., E.-K.C., E.J.B., E.D., E.S., E.Z.S., G.M.M., J.G.S., G.S., H.-N.P., H.A., H.B., H.H., H.J.C., H.J.L., H.M., H.S., I.E.C., I.S., J.B., J.E.K., J.G., J.I.R., J.P.K., J.P.P., J.S., J.T.R., J.v.M., K.C., K.H., K.I., K.L.L., K.S., L.-C.W., L.J.L., L.L., L.M.M., L.R., L.X., L.Z., M.C., M. Dichgans, M. Dörr, M.E.K., M.F., M.F.S., J.M.G., M.C.H., M.J.C., M.K., M.K.C., M.M.-N., M.P., M.R., M.S.O., M.S.S., M.T.-L., N.A.M., N.A.N., N.C., N.L.P., N.L.S., N.S., O.B.P., O.M., P.-S.Y., P.K., P.K.E.M., P.B.M., P.T.E., P.v.d.H., P.W.W., P.W.M., R.A.J.S., R.J., R.J.F.L., R.M., R.B.T., S.K.G., S.H.C., S.H.S., S.J.J., S. Kääb, S. Kany, S. Khurshid, S. Knight, S.A.L., S.M., S.M.D., S.N., S.R.O., S.P., S.R.H., S.W., S.Z., T.C., T.D., T.E., T.Z., U.T., V.G., J.W.J., W.M., X.G., X.Z. and Y.V.S. contributed to or revised the manuscript. B.W., C.R., D.G., E.-K.C., F.G., F.K.K., G.E.M.M., G.S., H.L., I.S., J.A.S., J.Y., K.M., L.-C.W., M.C., M.F., M.P., S.E.G., S.J.J., S.T., T.D. and W.Z. performed statistical analyses. A.A., C.D.A., C.G., C. Haggerty, C.M.A., C.T.R., C.W., D.C., D.I.C., D.L., D.M.R., D.S., E.J.B., E.B., F.G., H.-N.P., H.B., I.K., J.D.F., K.I., K.L.L., K.S., L.L., L.R., M.B.S., M.F., M.M.-N., M.P., M.S.S., N.A.M., P.M.R., P.T.E., Q.S.W., R.J.F.L., S.A.L., S.L.R.K. and Z.T.Y. contributed or managed samples and phenotype data. C.M.A., C.T.R., C.W., D.G., D.I.C., H.B., H.H., K.S. and P.T.E. conceived, designed and supervised the overall project.

Corresponding author

Correspondence to Patrick T. Ellinor.

Ethics declarations

Competing interests

B.M.P. serves on the steering committee of the Yale Open Data Access Project funded by Johnson & Johnson. C.D.A. receives sponsored research support from Bayer AG and has consulted for ApoPharma. C. Haggerty receives research support from Tempus Labs, outside the scope of the present work. C.M.A. receives sponsored research support from St. Jude, Abbott and Roche. C.R. is supported by a grant from Bayer AG to the Broad Institute focused on the development of therapeutics for cardiovascular disease. C.R. is a full-time employee of GSK as of 1 July 2024. C.T.R. reports research grants through Brigham and Women’s Hospital from Amgen, Anthos, AstraZeneca, Daiichi Sankyo, Janssen, Merck and Novartis and has received honoraria for scientific advisory boards and consulting from Anthos, Bayer, Bristol Myers Squibb, Daiichi Sankyo, Janssen, Pfizer, Regeneron and Sirius. The spouse of C.W. works at Regeneron Pharmaceuticals. D.O.A., D.G., G. Sveinbjornsson, H.A., H.H., K.S., R.B.T. and U.T. are employees of deCODE genetics/Amgen. D.C. has received consultancy fees from Roche Diagnostics and Trimedics, and speaker’s fees from BMS/Pfizer and Servier, all outside of the current work. D.F.F. is a full-time employee of Bayer. E.B. performs uncompensated consultancies and lectures with The Medicines Company. E.-K.C. reports research grants or speaking fees from Abbott, Bayer, BMS/Pfizer, Biosense Webster, Chong Kun Dang, Daewoong Pharmaceutical, Daiichi Sankyo, DeepQure, Dreamtech, Jeil Pharmaceutical, Medtronic, Samjinpharm, Seers Technology and Skylabs. L.-C.W. receives sponsored research support from IBM to the Broad Institute. L.Z. is a full-time employee of Bayer AG. M.E.K. is employed by SYNLAB Holding Deutschland. M.S.S. receives research grant support through Brigham and Women’s Hospital from Abbott, Amgen, Anthos Therapeutics, AstraZeneca, Bayer, Daiichi Sankyo, Eisai, Intarcia, Ionis, Medicines Company, MedImmune, Merck, Novartis, Pfizer and Quark Pharmaceuticals and consults for Althera, Amgen, Anthos Therapeutics, AstraZeneca, Beren Therapeutics, Bristol Myers Squibb, DalCor, Dr. Reddy’s Laboratories, Fibrogen, Intarcia, Merck, Moderna, Novo Nordisk and Silence Therapeutics; additionally, M.S.S. is a member of the TIMI Study Group, which has also received institutional research grant support through Brigham and Women’s Hospital from ARCA Biopharma, Janssen Research and Development, Siemens Healthcare Diagnostics, Softcell Medical Limited, Regeneron, Roche and Zora Biosciences. N.A.M. reports involvement in clinical trials with Amgen, Pfizer, Ionis, Novartis and AstraZeneca without personal fees, payments or increase in salary. P.M.R. has received investigator-initiated research grant support for unrelated projects from NHLBI, Operation Warp Speed, Novartis, Kowa, Amarin and Pfizer and has served as a consultant on unrelated issues to Novo Nordisk, Flame, Agepha, Uppton, Novartis, Jansen, Health Outlook, Civi Biopharm, Alnylam and SOCAR. P.T.E. receives sponsored research support from Bayer AG, Bristol Myers Squibb, Pfizer and Novo Nordisk; he has also served on advisory boards or consulted for Bayer AG. S.A.L. is a full-time employee of Novartis Institutes for BioMedical Research as of 18 July 2022. Previously, S.A.L. received sponsored research support from Bristol Myers Squibb/Pfizer, Bayer, Boehringer Ingelheim, Fitbit, IBM, Medtronic and Premier, and consulted for Bristol Myers Squibb/Pfizer, Bayer, Blackstone Life Sciences and Invitae. S.M.D. receives research support from RenalytixAI and personal consulting fees from Calico Labs, outside the scope of the current research. W.M. reports grants and/or personal fees from Siemens Healthineers, Aegerion Pharmaceuticals, AMGEN, AstraZeneca, Sanofi, Alexion Pharmaceuticals, BASF, Abbott Diagnostics, Numares, Berlin-Chemie, Akzea Therapeutics, Bayer Vital, bestbion dx, Boehringer Ingelheim Pharma, Immundiagnostik, Merck Chemicals, MSD Sharp and Dohme, Novartis Pharma and Olink Proteomics, and other from Synlab Holding Deutschland, all outside the submitted work. A.N. is a consultant for Abbott, Biosrnse Webster, Biotronik, Boston Sci, iRhythm, Field Medical, Pulse Bioselect and Medtronic. S. Khurshid receives sponsored research support from Bayer AG. All other authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Rafik Tadros and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Evaluation of effect estimates and allele frequencies for main GWAS meta-analysis and, in comparison, to the validation analysis.

a,b, Plots showing data for the 299 sentinel variants from the common variant analysis (MAF ≥ 1%) that were also available in the validation set. a, Correlation of allele frequencies between the meta-analysis and the validation cohort from MVP. b, Correlation of effect estimates between meta-analysis and the validation cohort from MVP. The red line is the identity line (x = y). Labelled in red are the variants with discordant direction of effect between meta-analysis and validation. c, Plot showing the relationship between effect allele frequency and strength of effect for sentinel variants of the meta-analysis. The effect estimates are from the inverse variance weighted method for meta-analysis. The dotted vertical lines show the cutoff for rare variants with MAF < 1%. The genome-wide significance cut-off of P < 5 × 10−8 was applied to correct for multiple testing. d, Plot showing co-occurrence of risk allele for atrial fibrillation and minor allele in blue and the inverse in red for sentinel variants of the meta-analysis. AF, allele frequency; ALL, all-ancestry; MAF, minor allele frequency; Meta, meta-analysis; MVP, Million Veteran Program.

Extended Data Fig. 2 Distribution of PoPS-score.

Red line shows the cutoff for mean + 3 standard deviations of the score (cutoff = 0.8548401). There were 205 genes with a PoPS-score higher than the cutoff.

Extended Data Fig. 3 Partitioned heritability analyses with annotations from human cardiac single-nuclei RNA-sequencing expression data.

a, Plot showing the partitioned heritability results for gene expression of cardiomyocytes by heart chamber (PLeftAtrium = 2.46 × 10−2, PRightAtrium = 1.51 × 10−1, PLeftVentricle = 6.25 × 10−1, PRightVentricle = 1.43 × 10−1). The blue dotted line shows the cutoff for nominal significance P < 0.05. The red dotted line shows the Bonferroni corrected significance cut off 0.05/4. b, Plot showing the partitioned heritability results for gene expression in left atrial tissue by cell type (PCardiomyocyte = 2.26 × 10−3, PFibroblast = 8.18 × 10−1, PEndothelial = 6.07 × 10−1, PPericyte = 4.34 × 10−2, PMacrophage = 3.48 × 10−1, PVSMC = 3.15 × 10−2, PAdipocyte = 8.90 × 10−1, PNeuronal = 1.56 × 10−1, PLymphocyte = 5.63 × 10−1). The blue dotted line shows the cutoff for nominal significance P < 0.05. The red dotted line shows the Bonferroni corrected significance cut off 0.05/9. VSMC, vascular smooth muscle cells.

Extended Data Fig. 4 Venn diagram of consensus genes between GenePrio and nearest protein coding genes.

Venn diagram for the overlap of GenePrio vs. nearest gene (protein coding, in relation to the transcription start position) at 139 loci. In red are the genes identified as the nearest, in blue are the GenePrio genes, and in black are the genes that overlap between the two groups. 56% of genes overlap.

Extended Data Fig. 5 Heatmap for GenePrio genes with two lines of evidence.

GenePrio genes with two lines of evidence. The five categories of evidence that were assessed to prioritize genes at GWAS loci: snRNA-seq (labelled as snRNA), gene was a top 10% marker gene for cardiomyocytes in left atrial tissue; Coding, gene had genome-wide significant loss-of-function variant or missense variant with predicted to be damaging effect; MAGMA, significant result for the gene in MAGMA analysis; PoPs, gene had a high PoPs score; eQTL, sentinel variant at locus had a significant eQTL to that gene in cardiac tissue. The genes are sorted from lowest to highest P-value at the sentinel variant of the locus. AF, atrial fibrillation; eQTL, expression quantitative trait locus; GWAS, genome-wide association study; MAF, minor allele frequency; MAGMA, Multi-marker Analysis of GenoMic Annotation; PoPS, polygenic priority score; snRNA-seq, single-nuclei RNA-sequencing.

Extended Data Fig. 6 Gene set enrichment analysis for all GenePrio genes.

Results of the gene set enrichment analysis for all 139 GenePrio genes across several databases. The −log10(P-values) are plotted sorted by gene set category. The top 5 gene sets by P-value are listed for each category. The size of each dot is proportional to the term size (n genes) of the gene set, that is larger terms have larger dots. The enrichment testing is done using a Fisher’s one-sided test (cumulative hypergeometric probability). P-values were adjusted for multiple testing using the g:SCS algorithm from the g:Profiler tool. BP, biological process; CC, cellular component; GO, gene ontology; HP, human phenotype ontology; HPA, human protein atlas; MF, molecular function; REAC, reactome; WP, wiki pathways.

Extended Data Fig. 7 Gene set enrichment results for top 10 GenePrio genes and GO:BP.

Heatmap of significant (adjusted P < 5 × 10−6) gene sets for GO:BP, showing top 10 GenePrio genes (GenePrio sum = 4) and their affiliation to each set. The enrichment testing is done using a Fisher’s one-sided test (cumulative hypergeometric probability). P-values were adjusted for multiple testing using the g:SCS algorithm from the g:Profiler tool. BP, biological process; GO, gene ontology.

Extended Data Fig. 8 Gene set enrichment results for top 10 GenePrio genes and GO:MF, GO:CC, HP, HPA and REAC.

Heatmap of significant (adjusted P < 5 × 10−6) gene sets for GO:MF, GO:CC, HP, HPA and REAC, showing top 10 GenePrio genes (GenePrio sum = 4) and their affiliation to each set. The enrichment testing is done using a Fisher’s one-sided test (cumulative hypergeometric probability). P-values were adjusted for multiple testing using the g:SCS algorithm from the g:Profiler tool. CC, cellular component; HP, human phenotype ontology; HPA, human protein atlas; MF, molecular function; REAC, Reactome.

Extended Data Fig. 9 Cluster analysis of the GenePrio genes based on cell type specific expression and Gene Ontology.

Results from a cluster analysis of 112 of the 139 GenePrio genes for Gene Ontology (GO) with clusterProfiler. Genes were annotated to cell types (one or more) based on the top 10% specific genes for each cell type. The over representation analysis uses a one-sided Fisher’s exact test. Multiple testing adjustment is performed with the Benjamini-Hochberg method. GO, gene ontology; VSMCs, vascular smooth muscle cells.

Extended Data Fig. 10 ATAC-seq tracks for TBX5, PITX2 and HSPB7 loci.

a, TBX5 locus. b, PITX2 locus. c, HSPB7 locus. Tracks for ATAC-seq from our iPSC-derived atrial cardiomyocytes and seven publicly available ENCODE ATAC-seq datasets (GM23338, liver, skeletal muscle, NK cells, pancreas, thyroid and bladder), as well as two histone modification tracks: H3K4me3 from a CUT&RUN experiment on our iPSC-derived atrial cardiomyocytes and layered H3K27ac from seven published ENCODE cell lines. The histone modifications H3K27ac and H3K4me3 are both associated with active regions in the genome, and are often located at promoters, enhancers or transcription start sites. GWAS variants indicate the ___location of common variants with genome-wide significance (P < 5 × 10−8) in the meta-analysis. Coordinates are in build GRCh38. ATAC-seq, Assay for Transposase-Accessible Chromatin using sequencing; CMs, cardiomyocytes; CUT&RUN, Cleavage Under Targets & Release Using Nuclease; ENCODE, Encyclopedia of DNA Elements; H3K27ac, histone H3 Lysine 27 acetylation; GWAS, genome-wide association study, H3K4me3, histone H3 lysine 4 trimethylation; iPS cells, induced pluripotent stem cells.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roselli, C., Surakka, I., Olesen, M.S. et al. Meta-analysis of genome-wide associations and polygenic risk prediction for atrial fibrillation in more than 180,000 cases. Nat Genet 57, 539–547 (2025). https://doi.org/10.1038/s41588-024-02072-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-024-02072-3

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