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.

Association analyses based on false discovery rate implicate new loci for coronary artery disease

Abstract

Genome-wide association studies (GWAS) in coronary artery disease (CAD) had identified 66 loci at 'genome-wide significance' (P < 5 × 10−8) at the time of this analysis, but a much larger number of putative loci at a false discovery rate (FDR) of 5% (refs. 1,2,3,4). Here we leverage an interim release of UK Biobank (UKBB) data to evaluate the validity of the FDR approach. We tested a CAD phenotype inclusive of angina (SOFT; ncases = 10,801) as well as a stricter definition without angina (HARD; ncases = 6,482) and selected cases with the former phenotype to conduct a meta-analysis using the two most recent CAD GWAS2,3. This approach identified 13 new loci at genome-wide significance, 12 of which were on our previous list of loci meeting the 5% FDR threshold2, thus providing strong support that the remaining loci identified by FDR represent genuine signals. The 304 independent variants associated at 5% FDR in this study explain 21.2% of CAD heritability and identify 243 loci that implicate pathways in blood vessel morphogenesis as well as lipid metabolism, nitric oxide signaling and inflammation.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Description of HARD and SOFT CAD phenotypes in UK Biobank.
Figure 2: Transposed Manhattan plot showing meta-analysis results for the SOFT CAD definition under an additive model.
Figure 3: Comparison of single-marker P values for the 5% FDR variants in the published CARDIoGRAMplusC4D 1000 Genomes–imputed CAD GWAS meta-analysis and the current FDR-based analysis.
Figure 4: Heat map showing DEPICT gene set enrichment results with zoom-in on a subset of the results.

Similar content being viewed by others

References

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

  2. Nikpay, M. et al. A comprehensive 1000 Genomes–based genome-wide association meta-analysis of coronary artery disease. Nat. Genet. 47, 1121–1130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Myocardial Infarction Genetics and CARDIoGRAM Exome Consortia Investigators. Coding variation in ANGPTL4, LPL, and SVEP1 and the risk of coronary disease. N. Engl. J. Med. 374, 1134–1144 (2016).

  4. Myocardial Infarction Genetics and CARDIoGRAM Exome Consortia Investigators. Systematic evaluation of pleiotropy identifies six further loci associated with coronary artery disease. J. Am. Coll. Cardiol. 69, 823–836 (2017).

  5. Marouli, E. et al. Rare and low-frequency coding variants alter human adult height. Nature 542, 186–190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Townsend, N., Bhatnagar, P., Wilkins, E., Wickramasinghe, K. & Rayner, M. Cardiovascular Disease Statistics 2015 (British Heart Foundation, 2015).

  7. Yang, J., Ferreira, T., Morris, A.P. & Medland, S.E. et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 44, 369–375 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Danesh, J. et al. EPIC-Heart: the cardiovascular component of a prospective study of nutritional, lifestyle and biological factors in 520,000 middle-aged participants from 10 European countries. Eur. J. Epidemiol. 22, 129–141 (2007).

    Article  PubMed  Google Scholar 

  9. Wipff, P.J., Rifkin, D.B., Meister, J.J. & Hinz, B. Myofibroblast contraction activates latent TGF-β1 from the extracellular matrix. J. Cell Biol. 179, 1311–1323 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Henderson, N.C. et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat. Med. 19, 1617–1624 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Goumans, M.J., Liu, Z. & ten Dijke, P. TGF-β signaling in vascular biology and dysfunction. Cell Res. 19, 116–127 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Nanda, V. et al. CDKN2B regulates TGFβ signaling and smooth muscle cell investment of hypoxic neovessels. Circ. Res. 118, 230–240 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Franzén, O. et al. Cardiometabolic risk loci share downstream cis- and trans-gene regulation across tissues and diseases. Science 353, 827–830 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Kukreja, R.C., Salloum, F.N. & Das, A. Cyclic guanosine monophosphate signaling and phosphodiesterase-5 inhibitors in cardioprotection. J. Am. Coll. Cardiol. 59, 1921–1927 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. The GTEx Consortium. The Genotype-Tissue Expression (GTEx) pilot analysis: multi-tissue gene regulation in humans. Science 348, 648–660 (2015).

  16. Samson, T. et al. The guanine-nucleotide exchange factor SGEF plays a crucial role in the formation of atherosclerosis. PLoS One 8, e55202 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Babaev, V.R. et al. Absence of regulated splicing of fibronectin EDA exon reduces atherosclerosis in mice. Atherosclerosis 197, 534–540 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Rohwedder, I. et al. Plasma fibronectin deficiency impedes atherosclerosis progression and fibrous cap formation. EMBO Mol. Med. 4, 564–576 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Fuchsberger, C. et al. The genetic architecture of type 2 diabetes. Nature 536, 41–47 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Surakka, I. et al. The impact of low-frequency and rare variants on lipid levels. Nat. Genet. 47, 589–597 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Howson, J.M.M. et al. Fifteen new risk loci for coronary artery disease highlight arterial-wall-specific mechanisms. Nat. Genet. 49, 1113–1119 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Pirinen, M., Donnelly, P. & Spencer, C.C. Including known covariates can reduce power to detect genetic effects in case–control studies. Nat. Genet. 44, 848–851 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Mägi, R. & Morris, A.P. GWAMA: software for genome-wide association meta-analysis. BMC Bioinformatics 11, 288 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Newson, R. QQVALUE: Stata module to generate quasi-q-values by inverting multiple-test procedures S457100 (Boston College Department of Economics, 2013).

  26. Bigdeli, T.B. et al. A simple yet accurate correction for winner's curse can predict signals discovered in much larger genome scans. Bioinformatics 32, 2598–2603 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Newson, R. Multiple-test procedures and smile plots. Stata J. 3, 109–132 (2003).

    Article  Google Scholar 

  28. Witte, J.S., Visscher, P.M. & Wray, N.R. The contribution of genetic variants to disease depends on the ruler. Nat. Rev. Genet. 15, 765–776 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Muñoz, M. et al. Evaluating the contribution of genetics and familial shared environment to common disease using the UK Biobank. Nat. Genet. 48, 980–983 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  30. 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).

    Article  CAS  PubMed  Google Scholar 

  31. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  32. Skipper, M. et al. Presenting the Epigenome Roadmap. Nature 518, 313 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Matys, V. et al. TRANSFAC: transcriptional regulation, from patterns to profiles. Nucleic Acids Res. 31, 374–378 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Portales-Casamar, E. et al. JASPAR 2010: the greatly expanded open-access database of transcription factor binding profiles. Nucleic Acids Res. 38, D105–D110 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Kircher, M. et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 46, 310–315 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ritchie, G.R., Dunham, I., Zeggini, E. & Flicek, P. Functional annotation of noncoding sequence variants. Nat. Methods 11, 294–296 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Boyle, A.P. et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 22, 1790–1797 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Staley, J.R. et al. PhenoScanner: a database of human genotype–phenotype associations. Bioinformatics 32, 3207–3209 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Fehrmann, R.S. et al. Gene expression analysis identifies global gene dosage sensitivity in cancer. Nat. Genet. 47, 115–125 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was funded by British Heart Foundation (BHF) grants RG/14/5/30893 to P.D. and FS/14/66/31293 to O.G. The work of P.D. forms part of the research themes contributing to the translational research portfolios of the Barts Biomedical Research Centre and Leicester Biomedical Research Centre funded by the UK National Institute for Health Research (NIHR). F.Y.L. and S.E.H. are funded by NIHR. C.P.N., T.R.W. and N.J.S. are funded from BHF, the Transatlantic Networks of Excellence Award (12CVD02) from the Leducq Foundation and EU-FP7/2007-2013 grant HEALTH-F2-2013-601456. N.J.S. is an NIHR Senior Investigator. PROCARDIS was supported by EU-FP6 (LSHM-CT- 2007-037273), AstraZeneca, BHF, the Swedish Research Council, the Knut and Alice Wallenberg Foundation, the Swedish Heart-Lung Foundation, the Torsten and Ragnar Söderberg Foundation, Karolinska Institutet, Foundation Strategic Research and the Stockholm County Council (560283). M.F. and H.W. are supported by Wellcome Trust award 090532/Z/09/Z, and M.F., H.W. and T.K. are supported by the BHF Centre of Research Excellence. A.G., H.W. and T.K. are supported by FP7/2007-2013 (HEALTH-F2-2013-601456 (CVGenes@Target)), and A.G. is supported by the Wellcome Trust and the TriPartite Immunometabolism Consortium-Novo Nordisk Foundation (NNF15CC0018486). HPS (ISRCTN48489393) was supported by the Medical Research Council (MRC), BHF, Merck and Co, and Roche Vitamins, Ltd. HPS acknowledges National Blood Service donor and UK-Twin Study controls (Wellcome Trust 07611, FP7/2007-2013). J.C.H. is funded by BHF (FS/14/55/30806). The Mount Sinai BioMe Biobank is supported by the Andrea and Charles Bronfman Philanthropies. The GLACIER Study and P.W.F. are funded by the European Commission (CoG-2015_681742_NASCENT), the Swedish Research Council (Distinguished Young Researchers Award), the Heart-Lung Foundation and the Novo Nordisk Foundation. OHGS studies were funded by the Canadian Institutes of Health Research, the Canada Foundation for Innovation and the Heart & Stroke Foundation of Canada. LURIC was funded from the EU-FP7 (Atheroremo (201668), RiskyCAD (305739), INTERREG IV Oberrhein Program), the European Regional Development Fund (ERDF), Wissenschaftsoffensive TMO and from the German Ministry for Education and Research, project e:AtheroSysMed (01ZX1313A-K). LOLIPOP is supported by the NIHR-BRC Imperial College Healthcare NHS Trust, BHF (SP/04/002), MRC (G0601966, G0700931), the Wellcome Trust (084723/Z/08/Z), NIHR (RP-PG-0407-10371), EU-FP7 (EpiMigrant, 279143) and Action on Hearing Loss (G51). The Helsinki Sudden Death Study was funded by EU-FP7 (201668, AtheroRemo), the Tampere University Foundation, Tampere University Hospital Medical Funds (grants 9M048 and 9N035 for T.L.), the Emil Aaltonen Foundation (T.L.), the Finnish Foundation of Cardiovascular Research (T.L., P.K.), the Pirkanmaa Regional Fund of the Finnish Cultural Foundation, the Yrjö Jahnsson Foundation, the Tampere Tuberculosis Foundation (T.L.), the Signe and Ane Gyllenberg Foundation (T.L.) and the Diabetes Research Foundation of the Finnish Diabetes Association (T.L.). M.T. (PG/16/49/32176) and R.C. (FS/12/80/29821) are supported by BHF. E.Z. acknowledges Wellcome Trust funding (098051). H.S. was supported by Deutsche Forschungsgemeinschaft (Sonderforschungsbereich CRC 1123 (B02)). The MRC/BHF Cardiovascular Epidemiology Unit was funded by MRC (G0800270), BHF (SP/09/002), NIHR-BRC Cambridge, the European Research Council (ERC; 268834), EU-FP7 (HEALTH-F2-2012-279233), Pfizer, Merck and Biogen. EPIC-CVD was supported by the University of Cambridge, EU-FP7 (HEALTH-F2-2012-279233), MRC (G0800270), BHF (SP/09/002) and ERC (268834). We thank all EPIC participants and staff and S. Spackman. EGCUT was funded by the Estonian Research Council grant for data management and the EPIC-CVD Coordinating Centre team. (IUT20-60), the Centre of Excellence in Genomics and Translational Medicine (GENTRANSMED), EU structural fund (Archimedes Foundation; 3.2.1001.11-0033), PerMed I and EU2020 (692145 ePerMed). This research was supported by BHF (SP/13/2/30111) and conducted using the UK Biobank Resource (application number 9922).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

C.P.N., A.G., A.S.B., S.K., T.R.W., E.M., I.N., J.C.H., O.G., H.S., M.F., J.D., N.J.S., H.W. and P.D. wrote and edited the manuscript. All authors contributed and discussed the results and commented on the manuscript. A.S.B., O.G., T.J., L.Z., S.E.H., E.A., T.L.A., E.P.B., J.C.C., R.C., R.M.C., P.E., R.E., E.E., P.W.F., C.G., D.G., A.H., J.M.M.H., E.I., A.K., T. Kessler, T. Kyriakou, T.L., X.L., Y.L., W.M., R.M., A.M., C.N.A.P., M.P.-R., A.F.S., M.J.S., P.A.Z., K.A., R.J.F.L., E.Z., J.E., G.D., H.S., J.D., N.J.S., H.W. and P.D. generated data and cohorts. C.P.N., A.S.B., I.N., F.Y.L., J.C.H., O.G., B.D.K., J.S.K., R.J.F.L., R.S.P., M.R., M.T., I.T., E.Z., J.E., G.D., H.S., J.D., N.J.S., H.W. and P.D. analyzed phenotype data for UKBB and replication studies. C.P.N., A.G., A.S.B., S.K., T.J. and M.F. performed the statistical analyses. C.P.N., S.K., T.R.W., A.S.B., R.E., A.R., E.E.S. and J.L.M.B. performed functional annotation. E.M. and P.D. performed biological and clinical enrichment and pathway analyses.

Corresponding author

Correspondence to Hugh Watkins.

Ethics declarations

Competing interests

P.W.F. has been a paid consultant for Eli Lilly and Sanofi Aventis and has received research support from several pharmaceutical companies as part of a European Union Innovative Medicines Initiative (IMI) project. E.I. is an advisor and consultant for Precision Wellness, Inc., and an advisor for Cellink for work unrelated to the present project. M.K.R. has acted as a consultant for GSK, Roche, Ascensia and MSD and participated in advisory board meetings on their behalf. M.K.R. has received lecture fees from MSD and grant support from Novo Nordisk, MSD and GSK. J.L.M.B. is the founder and chairman of Clinical Gene Networks. CGN has financially contributed to the STARNET study. J.L.M.B., E.E.S. and A.R. are on the board of directors for CGN. J.L.M.B. and A.R. own equity in CGN and receive financial compensation from CGN.

Additional information

A list of members and affiliations appears in the Supplementary Note.

A list of members and affiliations appears in the Supplementary Note.

A list of members and affiliations appears in the Supplementary Note.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10, Supplementary Tables 1, 3, 5, 8, 12 and 13, and Supplementary Note. (PDF 4854 kb)

Supplementary Tables 2, 4, 6, 7 and 9–11

Supplementary Tables 2, 4, 6, 7 and 9–11. (XLSX 265 kb)

Life Sciences Reporting Summary (PDF 160 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nelson, C., Goel, A., Butterworth, A. et al. Association analyses based on false discovery rate implicate new loci for coronary artery disease. Nat Genet 49, 1385–1391 (2017). https://doi.org/10.1038/ng.3913

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3913

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