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.

  • Article
  • Published:

Large-scale genome-wide association study of coronary artery disease in genetically diverse populations

Abstract

We report a genome-wide association study (GWAS) of coronary artery disease (CAD) incorporating nearly a quarter of a million cases, in which existing studies are integrated with data from cohorts of white, Black and Hispanic individuals from the Million Veteran Program. We document near equivalent heritability of CAD across multiple ancestral groups, identify 95 novel loci, including nine on the X chromosome, detect eight loci of genome-wide significance in Black and Hispanic individuals, and demonstrate that two common haplotypes at the 9p21 locus are responsible for risk stratification in all populations except those of African origin, in which these haplotypes are virtually absent. Moreover, in the largest GWAS for angiographically derived coronary atherosclerosis performed to date, we find 15 loci of genome-wide significance that robustly overlap with established loci for clinical CAD. Phenome-wide association analyses of novel loci and polygenic risk scores (PRSs) augment signals related to insulin resistance, extend pleiotropic associations of these loci to include smoking and family history, and precisely document the markedly reduced transferability of existing PRSs to Black individuals. Downstream integrative analyses reinforce the critical roles of vascular endothelial, fibroblast, and smooth muscle cells in CAD susceptibility, but also point to a shared biology between atherosclerosis and oncogenesis. This study highlights the value of diverse populations in further characterizing the genetic architecture of CAD.

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: Design of multi-population GWAS of CAD and estimates of heritability (h2) of CAD using GREML-LDMS-I for four populations.
Fig. 2: Population-specific GWAS and multi-population meta-analysis.
Fig. 3: Local ancestry and haplotype analyses at the 9p21 susceptibility locus for CAD in the MVP.
Fig. 4: Pleiotropic assessment of 95 novel loci through extended phenome-wide association of lead SNPs.
Fig. 5: Downstream analyses to prioritize systems, pathways, tissues and cells relevant to CAD.
Fig. 6: Testing of externally derived PRS and new multi-population scores in the MVP.

Similar content being viewed by others

Data availability

Summary statistics for the Biobank Japan study were obtained from http://jenger.riken.jp/en/result. Summary statistics for the CARDIoGRAMplusC4D study were obtained from http://www.cardiogramplusc4d.org. Summary statistics for the UK Biobank study for CAD were obtained from https://www.cardiomics.net/download-data. The full summary level association data from the individual population association analyses in MVP as well as the multi-population meta-analysis from this report will be available via the dbGaP Study accession number phs001672. This research has been conducted using the UK Biobank Resource under Application Numbers 13721 and 19416.

References

  1. Roth, G. A. et al. Global burden of cardiovascular diseases and risk factors, 1990–2019: update from the GBD 2019 Study. J. Am. Coll. Cardiol. 76, 2982–3021 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  2. National Center for Health Statistics. Health, United States Spotlight: Racial and Ethnic Disparities in Heart Disease (Centers for Disease Control and Prevention, 2019).

  3. Churchwell, K. et al. Call to action: structural racism as a fundamental driver of health disparities: a Presidential advisory from the American Heart Association. Circulation 142, e454–e468 (2020).

    PubMed  Google Scholar 

  4. Popejoy, A. B. & Fullerton, S. M. Genomics is failing on diversity. Nature 538, 161–164 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Martin, A. R. et al. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat. Genet. 51, 584–591 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Clarke, S. L., Assimes, T. L. & Tcheandjieu, C. The propagation of racial disparities in cardiovascular genomics research. Circ. Genom. Precis. Med. 14, e003178 (2021).

    Article  PubMed  Google Scholar 

  7. Zdravkovic, S. et al. Heritability of death from coronary heart disease: a 36-year follow-up of 20 966 Swedish twins. J. Intern. Med. 252, 247–254 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Wienke, A., Holm, N. V., Skytthe, A. & Yashin, A. I. The heritability of mortality due to heart diseases: a correlated frailty model applied to Danish twins. Twin Res. 4, 266–274 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. van der Harst, P. & Verweij, N. Identification of 64 novel genetic loci provides an expanded view on the genetic architecture of coronary artery disease. Circ. Res. 122, 433–443 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Koyama, S. et al. Population-specific and trans-ancestry genome-wide analyses identify distinct and shared genetic risk loci for coronary artery disease. Nat. Genet. 52, 1169–1177 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Webb, T. R. et al. Systematic evaluation of pleiotropy identifies 6 further loci associated with coronary artery disease. J. Am. Coll. Cardiol. 69, 823–836 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Coronary Artery Disease (C4D) Genetics Consortium. A genome-wide association study in Europeans and South Asians identifies five new loci for coronary artery disease. Nat. Genet. 43, 339–344 (2011).

    Article  CAS  Google Scholar 

  13. Lu, X. et al. Genome-wide association study in Han Chinese identifies four new susceptibility loci for coronary artery disease. Nat. Genet. 44, 890–894 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Assimes, T. L. & Roberts, R. Genetics: implications for prevention and management of coronary artery disease. J. Am. Coll. Cardiol. 68, 2797–2818 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Buniello, A. et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 47, D1005–D1012 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. National Center for Health Statistics (US). Crude percentages of all types of heart disease for adults aged 18 and over, United States, 2015-2018. National Health Interview Survey. Generated interactively: Wed Jul 13 2022.

  17. Institute for Health Metrics and Evaluation (IHME). GBD Compare Data Visualization (IHME, University of Washington, 2020). http://vizhub.healthdata.org/gbd-compare

  18. Nikpay, M. et al. A comprehensive 1,000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nat. Genet. 47, 1121–1130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ishigaki, K. et al. Large-scale genome-wide association study in a Japanese population identifies novel susceptibility loci across different diseases. Nat. Genet. 52, 669–679 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Barbalic, M. et al. Genome-wide association analysis of incident coronary heart disease (CHD) in African Americans: a short report. PLoS Genet. 7, e1002199 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lettre, G. et al. Genome-wide association study of coronary heart disease and its risk factors in 8,090 African Americans: the NHLBI CARe Project. PLoS Genet. 7, e1001300 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. Inouye, M. et al. Genomic risk prediction of coronary artery disease in 480,000 adults: implications for primary prevention. J. Am. Coll. Cardiol. 72, 1883–1893 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Dikilitas, O. et al. Predictive utility of polygenic risk scores for coronary heart disease in three major racial and ethnic groups. Am. J. Hum. Genet. 106, 707–716 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Speliotes, E. K. et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 42, 937–948 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Vujkovic, M. et al. Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat. Genet. 52, 680–691 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Liu, D. J. et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nat. Genet. 49, 1758–1766 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hartiala, J. A. et al. Genome-wide analysis identifies novel susceptibility loci for myocardial infarction. Eur. Heart J. 42, 919–933 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wainschtein, P. et al. Assessing the contribution of rare variants to complex trait heritability from whole-genome sequence data. Nat. Genet. 54, 263–273 (2022).

    Article  CAS  PubMed  Google Scholar 

  30. McPherson, R. et al. A common allele on chromosome 9 associated with coronary heart disease. Science 316, 1488–1491 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. CARDIoGRAMplusC4D Consortium et al. Large-scale association analysis identifies new risk loci for coronary artery disease. Nat. Genet. 45, 25–33 (2013).

    Article  CAS  Google Scholar 

  32. Shungin, D. et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature 518, 187–196 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Huang, Y. et al. Sexual differences in genetic predisposition of coronary artery disease. Circ. Genom. Precis. Med. 14, e003147 (2021).

    Article  CAS  PubMed  Google Scholar 

  34. Zore, T., Palafox, M. & Reue, K. Sex differences in obesity, lipid metabolism, and inflammation: a role for the sex chromosomes? Mol. Metab. 15, 35–44 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Salfati, E. et al. Susceptibility loci for clinical coronary artery disease and subclinical coronary atherosclerosis throughout the life-course. Circ. Cardiovasc. Genet. 8, 803–811 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Speliotes, E. K. et al. Genome-wide association analysis identifies variants associated with nonalcoholic fatty liver disease that have distinct effects on metabolic traits. PLoS Genet. 7, e1001324 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Natarajan, P. et al. Chromosome Xq23 is associated with lower atherogenic lipid concentrations and favorable cardiometabolic indices. Nat. Commun. 12, 2182 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Fletcher, R. et al. The role of the Niemann-Pick disease, type C1 protein in adipocyte insulin action. PLoS One 9, e95598 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Ghuran, A., van Der Wieken, L. R. & Nolan, J. Cardiovascular complications of recreational drugs. BMJ 323, 464–466 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wirka, R. C. et al. Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis. Nat. Med. 25, 1280–1289 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nelson, C. P. et al. Genetically determined height and coronary artery disease. N. Engl. J. Med. 372, 1608–1618 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ong, J. S. et al. Height and overall cancer risk and mortality: evidence from a Mendelian randomisation study on 310,000 UK Biobank participants. Br. J. Cancer 118, 1262–1267 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Clarke, S. L. et al. Broad clinical manifestations of polygenic risk for coronary artery disease in the Women’s Health Initiative. Preprint at https://doi.org/10.1101/2021.06.15.21258993 (2022).

  44. Xiao, B. et al. Inference of causal relationships based on the genetics of cardiometabolic traits and conditions unique to females in >50,000 participants. Preprint at https://doi.org/10.1101/2022.02.02.22269844 (2022).

  45. Singh, K. K. et al. BRCA1 is a novel target to improve endothelial dysfunction and retard atherosclerosis. J. Thorac. Cardiovasc. Surg. 146, 949–960 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Wu, H. T. et al. Oncogenic functions of the EMT-related transcription factor ZEB1 in breast cancer. J. Transl. Med. 18, 51 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ibrahim, N. et al. BRCA1-associated epigenetic regulation of p73 mediates an effector pathway for chemosensitivity in ovarian carcinoma. Cancer Res. 70, 7155–7165 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bai, F. et al. BRCA1 suppresses epithelial-to-mesenchymal transition and stem cell dedifferentiation during mammary and tumor development. Cancer Res. 74, 6161–6172 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Fardi, M., Alivand, M., Baradaran, B., Farshdousti Hagh, M. & Solali, S. The crucial role of ZEB2: from development to epithelial-to-mesenchymal transition and cancer complexity. J. Cell. Physiol. https://doi.org/10.1002/jcp.28277 (2019).

  50. Soini, Y. et al. Transcription factors zeb1, twist and snai1 in breast carcinoma. BMC Cancer 11, 73 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cheng, P. et al. ZEB2 shapes the epigenetic landscape of atherosclerosis. Circulation 145, 469–485 (2022).

    Article  CAS  PubMed  Google Scholar 

  52. Tabas, I., Garcia-Cardena, G. & Owens, G. K. Recent insights into the cellular biology of atherosclerosis. J. Cell Biol. 209, 13–22 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Nagao, M. et al. Coronary disease-associated gene TCF21 inhibits smooth muscle cell differentiation by blocking the myocardin-serum response factor pathway. Circ. Res. 126, 517–529 (2020).

    Article  CAS  PubMed  Google Scholar 

  54. Lowrie Jr, D. J. Histology: An Essential Textbook (Thieme Publishers, 2020).

  55. Ko, C. W., Qu, J., Black, D. D. & Tso, P. Regulation of intestinal lipid metabolism: current concepts and relevance to disease. Nat. Rev. Gastroenterol. Hepatol. 17, 169–183 (2020).

    Article  CAS  PubMed  Google Scholar 

  56. Fahed, A. C. et al. Transethnic transferability of a genome-wide polygenic score for coronary artery disease. Circ. Genom. Precis. Med. 14, e003092 (2021).

    Article  PubMed  Google Scholar 

  57. Gaziano, J. M. et al. Million Veteran Program: a mega-biobank to study genetic influences on health and disease. J. Clin. Epidemiol. 70, 214–223 (2016).

    Article  PubMed  Google Scholar 

  58. Hunter-Zinck, H. et al. Genotyping array design and data quality control in the Million Veteran Program. Am. J. Hum. Genet. 106, 535–548 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Genomes Project Consortiumet al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  CAS  Google Scholar 

  60. Loh, P. R. et al. Reference-based phasing using the Haplotype Reference Consortium panel. Nat. Genet. 48, 1443–1448 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Fang, H. et al. Harmonizing genetic ancestry and self-identified race/ethnicity in genome-wide association studies. Am. J. Hum. Genet. 105, 763–772 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Byrd, J. B. et al. Data quality of an electronic health record tool to support VA cardiac catheterization laboratory quality improvement: the VA Clinical Assessment, Reporting, and Tracking System for Cath Labs (CART) program. Am. Heart J. 165, 434–440 (2013).

    Article  PubMed  Google Scholar 

  64. Maddox, T. M. et al. A national clinical quality program for Veterans Affairs catheterization laboratories (from the Veterans Affairs clinical assessment, reporting, and tracking program). Am. J. Cardiol. 114, 1750–1757 (2014).

    Article  PubMed  Google Scholar 

  65. Maddox, T. M. et al. Nonobstructive coronary artery disease and risk of myocardial infarction. JAMA 312, 1754–1763 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Yang, J. et al. Genetic variance estimation with imputed variants finds negligible missing heritability for human height and body mass index. Nat. Genet. 47, 1114–1120 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Evans, L. M. et al. Comparison of methods that use whole genome data to estimate the heritability and genetic architecture of complex traits. Nat. Genet. 50, 737–745 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lee, S. H., Wray, N. R., Goddard, M. E. & Visscher, P. M. Estimating missing heritability for disease from genome-wide association studies. Am. J. Hum. Genet. 88, 294–305 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Lee, S. H. et al. Estimating the proportion of variation in susceptibility to schizophrenia captured by common SNPs. Nat. Genet. 44, 247–250 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Visscher, P. M. et al. Statistical power to detect genetic (co)variance of complex traits using SNP data in unrelated samples. PLoS Genet. 10, e1004269 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bulik-Sullivan, B. K. et al. LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat. Genet. 47, 291–295 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Magi, R. et al. Trans-ethnic meta-regression of genome-wide association studies accounting for ancestry increases power for discovery and improves fine-mapping resolution. Hum. Mol. Genet. 26, 3639–3650 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Loley, C. et al. No association of coronary artery disease with X-chromosomal variants in comprehensive international meta-analysis. Sci. Rep. 6, 35278 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Graham, S. E. et al. The power of genetic diversity in genome-wide association studies of lipids. Nature 600, 675–679 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Watanabe, K., Taskesen, E., van Bochoven, A. & Posthuma, D. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun. 8, 1826 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Coram, M. A. et al. Leveraging multi-ethnic evidence for mapping complex traits in minority populations: an empirical Bayes approach. Am. J. Hum. Genet. 96, 740–752 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Choi, S. W. & O’Reilly, P. F. PRSice-2: Polygenic Risk Score software for biobank-scale data. Gigascience 8, giz082 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Denny, J. C. et al. PheWAS: demonstrating the feasibility of a phenome-wide scan to discover gene–disease associations. Bioinformatics 26, 1205–1210 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wu, P. et al. Mapping ICD-10 and ICD-10-CM codes to phecodes: workflow development and initial evaluation. JMIR Med. Inform. 7, e14325 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  82. Global Lipids Genetics Consortiumet al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

    Article  CAS  Google Scholar 

  83. Evangelou, E. et al. Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat. Genet. 50, 1412–1425 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Erzurumluoglu, A. M. et al. Meta-analysis of up to 622,409 individuals identifies 40 novel smoking behaviour associated genetic loci. Mol. Psychiatry 25, 2392–2409 (2020).

    Article  CAS  PubMed  Google Scholar 

  85. Yengo, L. et al. Meta-analysis of genome-wide association studies for height and body mass index in approximately 700000 individuals of European ancestry. Hum. Mol. Genet. 27, 3641–3649 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Xue, A. et al. Genome-wide association analyses identify 143 risk variants and putative regulatory mechanisms for type 2 diabetes. Nat. Commun. 9, 2941 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Maples, B. K., Gravel, S., Kenny, E. E. & Bustamante, C. D. RFMix: a discriminative modeling approach for rapid and robust local-ancestry inference. Am. J. Hum. Genet. 93, 278–288 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. 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  CAS  Google Scholar 

  89. de Leeuw, C. A., Stringer, S., Dekkers, I. A., Heskes, T. & Posthuma, D. Conditional and interaction gene-set analysis reveals novel functional pathways for blood pressure. Nat. Commun. 9, 3768 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Zhu, X. & Stephens, M. Large-scale genome-wide enrichment analyses identify new trait-associated genes and pathways across 31 human phenotypes. Nat. Commun. 9, 4361 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Pers, T. H. et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat. Commun. 6, 5890 (2015).

    Article  CAS  PubMed  Google Scholar 

  92. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This research is based on data from the Million Veteran Program, Office of Research and Development, Veterans Health Administration, and was supported by Veterans Administration awards I01-01BX003362, I01-BX004821 (K.-M.C., P.S.T.), I01-BX003340 (K.Cho, P.W.F.W.) and VA HSR RES 13-457 (VA Informatics and Computing Infrastructure). The content of this manuscript does not represent the views of the Department of Veterans Affairs or the United States Government. The eMERGE Network was initiated and funded by the National Human Genome Research Institute (NHGRI) through the following grants: Phase III: U01HG8657 (Kaiser Permanente Washington/University of Washington); U01HG8685 (Brigham and Women’s Hospital); U01HG8672 (Vanderbilt University Medical Center); U01HG8666 (Cincinnati Children’s Hospital Medical Center); U01HG6379 (Mayo Clinic); U01HG8679 (Geisinger Clinic); U01HG8680 (Columbia University Health Sciences); U01HG8684 (Children’s Hospital of Philadelphia); U01HG8673 (Northwestern University); U01HG8701 (Vanderbilt University Medical Center serving as the Coordinating Center); U01HG8676 (Partners Healthcare/Broad Institute); and U01HG8664 (Baylor College of Medicine); Phase II: U01HG006828 (Cincinnati Children’s Hospital Medical Center/Boston Children’s Hospital); U01HG006830 (Children’s Hospital of Philadelphia); U01HG006389 (Essentia Institute of Rural Health, Marshfield Clinic Research Foundation and Pennsylvania State University); U01HG006382 (Geisinger Clinic); U01HG006375 (Group Health Cooperative/University of Washington); U01HG006379 (Mayo Clinic); U01HG006380 (Icahn School of Medicine at Mount Sinai); U01HG006388 (Northwestern University); U01HG006378 (Vanderbilt University Medical Center); and U01HG006385 (Vanderbilt University Medical Center serving as the Coordinating Center). Phase II: U01HG004438 (CIDR) and U01HG004424 (the Broad Institute) serving as Genotyping Centers. Phase I: U01-HG-004610 (Group Health Cooperative/University of Washington); U01-HG-004608 (Marshfield Clinic Research Foundation and Vanderbilt University Medical Center); U01-HG-04599 (Mayo Clinic); U01HG004609 (Northwestern University); U01-HG-04603 (Vanderbilt University Medical Center, also serving as the Administrative Coordinating Center); U01HG004438 (CIDR) and U01HG004424 (the Broad Institute) serving as Genotyping Centers. The Population Architecture Using Genomics and Epidemiology (PAGE) program is funded by the NHGRI with co-funding from the National Institute on Minority Health and Health Disparities (NIMHD), supported by U01HG007416 (CALiCo), U01HG007417 (ISMMS), U01HG007397 (MEC), U01HG007376 (WHI), and U01HG007419 (Coordinating Center). The MultiEthnic Study (MEC) was supported by U01 CA164973. The Women’s Health Initiative (WHI) program is funded by the National Heart, Lung, and Blood Institute, National Institutes of Health, US Department of Health and Human Services through contracts HHSN268201100046C, HHSN268201100001C, HHSN268201100002C, HHSN268201100003C, HHSN268201100004C and HHSN271201100004C. Scientific Computing Infrastructure at Fred Hutch is funded by ORIP grant S10OD028685. Funding support for the ‘Exonic variants and their relation to complex traits in minorities of the WHI study is provided through the NHGRI PAGE program (U01HG004790). The Atherosclerosis Risk in Communities (ARIC) Study is carried out as a collaborative study supported by National Heart, Lung, and Blood Institute contracts (HHSN268201100005C, HHSN268201100006C, HHSN268201100007C, HHSN268201100008C, HHSN268201100009C, HHSN268201100010C, HHSN268201100011C and HHSN268201100012C). The Cardiovascular Health Study (CHS) was supported by NHLBI contracts HHSN268201200036C, HHSN268200800007C, HHSN268201800001C, N01HC55222, N01HC85079, N01HC85080, N01HC85081, N01HC85082, N01HC85083, N01HC85086, 75N92021D00006; and NHLBI grants U01HL080295, R01HL085251, R01HL087652, R01HL105756, R01HL103612, R01HL120393 and U01HL130114 with additional contribution from the National Institute of Neurological Disorders and Stroke (NINDS). Additional support was provided through R01AG023629 from the National Institute on Aging (NIA). A full list of principal CHS investigators and institutions can be found at CHS-NHLBI.org. The provision of genotyping data was supported in part by the National Center for Advancing Translational Sciences, CTSI grant UL1TR001881, and the National Institute of Diabetes and Digestive and Kidney Disease Diabetes Research Center (DRC) grant DK063491 to the Southern California Diabetes Endocrinology Research Center. BioBank Japan (BBJ) was supported by the Tailor-Made Medical Treatment Program of the Ministry of Education, Culture, Sports, Science, and Technology and Japan Agency for Medical Research (AMED) under grant numbers JP17km0305002 and JP17km0305001. Healthy Aging in Neighborhoods of Diversity across the Life Span (HANDLS) was funded by the Interlaboratory Proposal Funding of the Intramural Research Program of the National Institute on Aging (NIA), the National Institutes of Health (NIH), Baltimore, Maryland. Funding number: [AG000989]. X.Z. was supported by the Stein Fellowship from Stanford University and Institute for Computational and Data Sciences Seed Grant from The Pennsylvania State University. S.M.D., J.A.L. and K.M.L. were supported by the US Department of Veterans Affairs (IK2-CX001780). Y.L. is supported by NIH R56HL150186. S.Ko. and K.It. were supported by AMED under Grant Numbers JP20km0405209 and JP20ek0109487. K.E.N. is supported by NIH R01HL142302. R.D. is supported by NIH R35GM124836 and R01HL139865. F.C. is supported by NCI T32CA229110. B.F.V. was supported by the NIH R01DK101478 and a Linda Pechenik Montague Investigator Award. P.N. is supported by grants from the NIH/NHLBI (R01HL142711, R01HL148050, R01HL127564, R01HL151152), NIH/NHGRI (U01HG011719), Fondation Leducq (TNE-18CVD04) and Massachusetts General Hospital (Fireman Chair). Support for title page creation and format was provided by AuthorArranger, a tool developed at the National Cancer Institute. The authors thank C. D. Bustamante for his review and feedback of specific cross-population analyses involving the 9p21 region. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

Concept and design: C.T., X.Z., A.T.H., S.L.C., V.N., D.J.R., K.-M.C., J.A.L., S.M.D., P.W.F.W., H.T., Y.V.S., P.S.T., C.J.O., T.L.A. Acquisition, analysis or interpretation of data: C.T., X.Z., A.T.H., S.L.C., V.N., S.M., B.R.G., K.M.L., R.S., K.M.K., H.F., F.C., Y.L., S.Ko., N.L.T., M.Vu., S.R., M.E.P., T.M.M., S.W.W., A.G.B., M.G.L., S.P., J.Hu., N.S.-A., Y.-L.H., G.L.W., S.B., C.K., J.Ha., R.J.F.L., R.D., M.Ve., K.Cha., K.E.N., C.L.A., M.G., C.A.H., L.L.M., L.R.W., J.C.B., H.L., B.S., L.A.La., A.G., O.D., I.J.K., I.B.S., G.P.J., A.S.G., S.H., B.N., K.It., K.Is., Y.K., S.S.V., M.D.R., R.L.K., A.B., L.A.Lo., S.Ka., E.R.H., D.R.M., J.S.L., D.S., P.D.R., K.Cho, J.M.G., J.E.H., B.F.V., D.J.R., K.-M.C., J.A.L., S.M.D., P.W.F.W, H.T., Y.V.S., P.S.T., C.J.O., T.L.A. Drafting of the manuscript: C.T., T.L.A. Critical revision of the manuscript for important intellectual content: X.Z., A.T.H., S.L.C., V.N., M.Vu., D.K., S.R., M.G.L., R.D., K.E.N., C.K., J.C.B., I.J.K., M.D.R., P.N., B.F.V., J.A.L., S.M.D., P.W.F.W, H.T., Y.V.S., P.S.T, C.J.O.

Corresponding authors

Correspondence to Catherine Tcheandjieu or Themistocles L. Assimes.

Ethics declarations

Competing interests

A.B. and L.A.Lo. are employees of Regeneron Pharmaceuticals. R.D. has received grants from AstraZeneca, grants and non-financial support from Goldfinch Bio, is a scientific co-founder, consultant and equity holder for Pensieve Health and a consultant for Variant Bio. T.M.M. is an employee of the Healthcare Innovation Lab at BJC HealthCare/Washington University School of Medicine, an advisor of Myia Labs, and a compensated director of the JF Maddox Foundation in New Mexico. S.Ka. is an employee of Verve Therapeutics, holds equity in Verve Therapeutics and Maze Therapeutics, and has served as a consultant for Acceleron, Eli Lilly, Novartis, Merck, Novo Nordisk, Novo Ventures, Ionis, Alnylam, Aegerion, Haug Partners, Noble Insights, Leerink Partners, Bayer Healthcare, Illumina, Color Genomics, MedGenome, Quest and Medscape. D.J.R. is on the Scientific Advisory Board of Alnylam, Novartis and Verve Therapeutics. M.D.R. is on the scientific advisory board for Goldfinch Bio and Cipherome. C.J.O. became an employee of Novartis after the initial submission of the manuscript. P.N. reports investigator-initiated grants from Amgen, Apple, AstraZeneca, Boston Scientific and Novartis, personal fees from Apple, AstraZeneca, Blackstone Life Sciences, Invitae, Foresite Labs, Novartis and Roche/Genentech, and is a co-founder of TenSixteen Bio, a shareholder of geneXwell, TenSixteen Bio and Vertex, a scientific advisory board member of geneXwell and TenSixteen Bio, and reports spousal employment at Vertex, all unrelated to the present work. S.M.D. receives research support from RenalytixAI to his institution and consulting fees from Calico Labs. A.G.B. is a scientific co-founder and equity holder in TenSixteen Bio. All other authors have no competing interests.

Peer review

Peer review information

Nature Medicine thanks Riyaz Patel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Michael Basson, in collaboration with the Nature Medicine team.

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 LocusZoom plots of loci reaching genome-wide significance in Black participants and Hispanic participants.

Sets of LocusZoom plots for five loci in Black participants and 3 loci in Hispanic participants reaching genome-wide significance after two-stage meta-analysis with external cohorts. Each set of plots show the association results for a locus for all three populations using the same chromosome location scale (x-axis) but not the same p-value scale (y-axis). P values are derived from inverse variance-weighted meta-analysis using METAL and are two-sided.

Extended Data Fig. 2 Allele frequencies and association results at the 9p21 locus among Black in the Million Veteran Program stratified by local ancestry status.

Top panels show plots of corresponding allelic frequencies at the 9p21 susceptibility locus observed in MVP white participants vs. subgroups of MVP Black participants with a. two African chromosomes (chr), b. one African chr, and c. no African chr at the locus. Corresponding LocusZoom plots for each group are in the panels immediately below. Association testing was performed using logistic regression with adjustment on sex and principal component as implemented in PLINK. P values were derived from a Wald test and are two-sided.

Extended Data Fig. 3 LocusZoom plots of SNP association at the 9p21 susceptibility locus for CAD.

Top panel plots the results for MVP GWAS of all Hispanic participants + Stage 2 cohort meta-analysis. P values are derived from inverse variance weighted meta-analysis using METAL and are two-sided. Bottom panel plots the subset of MVP Hispanic participants with no African derived chromosomes at 9p21 based on local ancestry assessment using RFMix (5,298 cases/20,556 controls). Association testing was performed using logistic regression with adjustment on sex and principal component as implemented in PLINK. P values were derived from a Wald test and are two-sided.

Extended Data Table 1 Demographic characteristics of the Million Veteran Program participants included in the genome-wide association study analyses of the clinical coronary artery disease phenotype. SD: standard deviation. AMI: acute myocardial infarction. HARE: Harmonizing Genetic Ancestry and Self-identified Race/Ethnicity algorithm. CAD: coronary artery disease
Extended Data Table 2 Demographic characteristics of the white, Black, and Hispanic participants from the Million Veteran Program and the Japanese from Biobank Japan used in GREML-LDMS-I heritability analyses. N: number in stratum. SD: standard deviation
Extended Data Table 3 Characteristics of Stage 1 and Stage 2 Black and Hispanic cohorts. PAGE: Population Architecture through Genomics and Environment Study (funded by the National Institutes of Health - NHGRI). ARIC: Atherosclerosis Risk in Communities Study (funded by the National institutes of Health - NHLBI). MEC: Multiethnic Study (funded by the National Cancer Institute). WHI: Women’s Health Initiative study (funded by the National institutes of Health - NHLBI). MEGA (WHI): PAGE substudy in the WHI genotyped with the Illumina Multi-Ethnic Genotyping Array. SHARE: SNP Health Association Resource substudy in WHI genotyped using Affymetrix 6.0 array. GARNET: Genomics and Randomized Trials Network substudy in WHI genotyped using Illumina HumanOmni1-Quad v1-0 B. BioVU: Vanderbilt’s biorepository of DNA extracted from discarded blood collected during routine clinical testing and linked to de-identified medical records in the Synthetic Derivative. CHS: Cardiovascular Health Study (funded by the National institutes of Health - NHLBI). Health ABC: The Health, Aging and Body Composition Study (funded by the National institutes of Health - NIA). HANDLS: The Healthy Aging in Neighborhoods of Diversity across the Life Span study (funded by the National Institutes of Health - NIA). JHS: The Jackson Heart Study (funded by the National Institutes of Health - NHLBI and NIMHD). eMERGE: The electronics Medical records and Genomics consortium (funded by the National Institutes of Health - NHGRI). Penn Biobank: The Penn Medicine BioBank (Institute for Translational Medicine and Therapeutics at the University of Pennsylvania). UK-Biobank: The UK Biobank Study. BioME: The BioME Biobank Program (The Institute for Personalized Medicine at the Icahn School of Medicine at Mount Sinai)
Extended Data Table 4 Loci reaching genome-wide significance after two-stage meta-analysis in Black participants and Hispanic participants. *The 8p22 locus in Black participants was carefully examined for the possibility of a false-positive association as was not even a hint of a genetic signal in this region for white participants despite more than adequate power due to much larger sample size and a substantially higher frequency of the lead SNPs. There was no obvious link between the signal and the status of the inversion in the immediately adjacent 8p23 region which we called using principal component analyses of SNPs within the inversion site. The region reached genome-wide significance in MVP Black participants based solely on imputed genotypes. We determined that neighboring genotypes in the region were not reliably called due to an unrecognized African specific deletion in the region subsequently reported by the gnoMAD consortium. The deletion also affected the reliability of the imputed SNPs. Even after recalling the genotypes in this region taking into consideration the presence of the deletion, no genotyped SNPs were genome-wide significant for CAD. P values are derived from inverse variance weighted meta-analysis using METAL and are two-sided
Extended Data Table 5 Derivation of the VA Clinical Assessment Reporting and Tracking (CART) sub cohort for genome-wide association study. 1 V = 1 vessel >50% obstruction, 2 V = 2 vessels with >50% obstruction, 3 V/LM = 3 vessels and/or left main disease >50% obstruction. *age assigned to last procedure. **age assigned to earliest procedure. ***age assigned to the earliest procedure showing the most severe disease. ****n with history of a cardiac transplant = 137, major age discrepancy between CART and CDW derived age = 2, and undefined HARE assignment = 595
Extended Data Table 6 Age and sex characteristics of VA Clinical Assessment Reporting and Tracking (CART) subcohort by Harmonizing Genetic Ancestry and Self-identified Race/Ethnicity algorithm (HARE) assigned populations and severity of disease. *did not proceed with genetic association in this group because of low numbers
Extended Data Table 7 Odds ratio of CAD per standard deviation increase of the externally derived LDPred and metaGRS scores in the MVP white, Black, and Hispanic cohorts followed by relative efficiency estimates based on ratios of betas. *LDPred score for CAD as previously described22 **metaGRS for CAD as previously described23 ***AMI/Revasc: subset of cases with evidence of discharge diagnosis of acute myocardial infarction or revascularization procedure in the EHR. All CAD: further include participants with CAD codes that are not AMI or revascularization **** Relative efficiency: ratio of log ORs (beta coefficients) between MVP and UK Biobank. n/a: not applicable as this broader phenotype not reported in the LDpred and metaGRS reports. glm function in R for logistic regression covariates: age, sex, genotyping batch and top 10 genotype-based PCs. p-value corresponding to the z ratio based on a Standard Normal reference distribution, 2-sided

Supplementary information

Supplementary Information

Supplementary Methods and list of Consortia authors

Reporting Summary

Supplementary Table 1

Supplementary Tables 1–37 and list of abbreviations used in the Tables

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tcheandjieu, C., Zhu, X., Hilliard, A.T. et al. Large-scale genome-wide association study of coronary artery disease in genetically diverse populations. Nat Med 28, 1679–1692 (2022). https://doi.org/10.1038/s41591-022-01891-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-022-01891-3

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