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:

Exome-wide association study of plasma lipids in >300,000 individuals

Abstract

We screened variants on an exome-focused genotyping array in >300,000 participants (replication in >280,000 participants) and identified 444 independent variants in 250 loci significantly associated with total cholesterol (TC), high-density-lipoprotein cholesterol (HDL-C), low-density-lipoprotein cholesterol (LDL-C), and/or triglycerides (TG). At two loci (JAK2 and A1CF), experimental analysis in mice showed lipid changes consistent with the human data. We also found that: (i) beta-thalassemia trait carriers displayed lower TC and were protected from coronary artery disease (CAD); (ii) excluding the CETP locus, there was not a predictable relationship between plasma HDL-C and risk for age-related macular degeneration; (iii) only some mechanisms of lowering LDL-C appeared to increase risk for type 2 diabetes (T2D); and (iv) TG-lowering alleles involved in hepatic production of TG-rich lipoproteins (TM6SF2 and PNPLA3) tracked with higher liver fat, higher risk for T2D, and lower risk for CAD, whereas TG-lowering alleles involved in peripheral lipolysis (LPL and ANGPTL4) had no effect on liver fat but decreased risks for both T2D and 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

Figure 1: A1CF p.Gly398Ser mutant leads to increased APOB100 secretion.
Figure 2: Association of genetically lowered triglycerides by LPL variants with a range of phenotypes.

Similar content being viewed by others

References

  1. Teslovich, T.M. et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466, 707–713 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Asselbergs, F.W. et al. Large-scale gene-centric meta-analysis across 32 studies identifies multiple lipid loci. Am. J. Hum. Genet. 91, 823–838 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Willer, C.J. et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Albrechtsen, A. et al. Exome sequencing-driven discovery of coding polymorphisms associated with common metabolic phenotypes. Diabetologia 56, 298–310 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Peloso, G.M. et al. Association of low-frequency and rare coding-sequence variants with blood lipids and coronary heart disease in 56,000 whites and blacks. Am. J. Hum. Genet. 94, 223–232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. Tang, C.S. et al. Exome-wide association analysis reveals novel coding sequence variants associated with lipid traits in Chinese. Nat. Commun. 6, 10206 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Musunuru, K. et al. From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature 466, 714–719 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Burkhardt, R. et al. Trib1 is a lipid- and myocardial infarction-associated gene that regulates hepatic lipogenesis and VLDL production in mice. J. Clin. Invest. 120, 4410–4414 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Voight, B.F. et al. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet 380, 572–580 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Do, R. et al. Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat. Genet. 45, 1345–1352 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lu, X. et al. Exome chip meta-analysis identifies novel loci and East Asian–specific coding variants contributing to lipid levels and coronary artery disease. Nat. Genet. http://dx.doi.org/10.1038/ng.3978 (2017).

  13. Feng, S., Liu, D., Zhan, X., Wing, M.K. & Abecasis, G.R. RAREMETAL: fast and powerful meta-analysis for rare variants. Bioinformatics 30, 2828–2829 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Liu, D.J. et al. Meta-analysis of gene-level tests for rare variant association. Nat. Genet. 46, 200–204 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Locke, A.E. et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 518, 197–206 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Holmen, O.L. et al. Systematic evaluation of coding variation identifies a candidate causal variant in TM6SF2 influencing total cholesterol and myocardial infarction risk. Nat. Genet. 46, 345–351 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Shen, X. et al. Identification of genes affecting apolipoprotein B secretion following siRNA-mediated gene knockdown in primary human hepatocytes. Atherosclerosis 222, 154–157 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Baxter, E.J. et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365, 1054–1061 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. James, C. et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 434, 1144–1148 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Kralovics, R. et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N. Engl. J. Med. 352, 1779–1790 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Levine, R.L. et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7, 387–397 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Jaiswal, S. et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N. Engl. J. Med. 377, 111–121 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Mullally, A. et al. Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. Cancer Cell 17, 584–596 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lellek, H. et al. Purification and molecular cloning of a novel essential component of the apolipoprotein B mRNA editing enzyme-complex. J. Biol. Chem. 275, 19848–19856 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Mehta, A., Kinter, M.T., Sherman, N.E. & Driscoll, D.M. Molecular cloning of apobec-1 complementation factor, a novel RNA-binding protein involved in the editing of apolipoprotein B mRNA. Mol. Cell. Biol. 20, 1846–1854 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Galloway, C.A., Ashton, J., Sparks, J.D., Mooney, R.A. & Smith, H.C. Metabolic regulation of APOBEC-1 complementation factor trafficking in mouse models of obesity and its positive correlation with the expression of ApoB protein in hepatocytes. Biochim. Biophys. Acta 1802, 976–985 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Galanello, R. & Origa, R. Beta-thalassemia. Orphanet J. Rare Dis. 5, 11 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Fessas, P., Stamatoyannopoulos, G. & Keys, A. Serum-cholesterol and thalassemia trait. Lancet 1, 1182–1183 (1963).

    Article  CAS  PubMed  Google Scholar 

  30. Sidore, C. et al. Genome sequencing elucidates Sardinian genetic architecture and augments association analyses for lipid and blood inflammatory markers. Nat. Genet. 47, 1272–1281 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chen, W. et al. Genetic variants near TIMP3 and high-density lipoprotein-associated loci influence susceptibility to age-related macular degeneration. Proc. Natl. Acad. Sci. USA 107, 7401–7406 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Neale, B.M. et al. Genome-wide association study of advanced age-related macular degeneration identifies a role of the hepatic lipase gene (LIPC). Proc. Natl. Acad. Sci. USA 107, 7395–7400 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wang, Y.F. et al. CETP/LPL/LIPC gene polymorphisms and susceptibility to age-related macular degeneration. Sci. Rep. 5, 15711 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cheng, C.Y. et al. New loci and coding variants confer risk for age-related macular degeneration in East Asians. Nat. Commun. 6, 6063 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Momozawa, Y. et al. Low-frequency coding variants in CETP and CFB are associated with susceptibility of exudative age-related macular degeneration in the Japanese population. Hum. Mol. Genet. 25, 5027–5034 (2016).

    CAS  PubMed  Google Scholar 

  36. Lotta, L.A. et al. Association between low-density lipoprotein cholesterol-lowering genetic variants and risk of type 2 diabetes: A meta-analysis. J. Am. Med. Assoc. 316, 1383–1391 (2016).

    Article  CAS  Google Scholar 

  37. Schmidt, A.F. et al. PCSK9 genetic variants and risk of type 2 diabetes: a mendelian randomisation study. Lancet Diabetes Endocrinol. 5, 97–105 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ference, B.A. et al. Variation in PCSK9 and HMGCR and risk of cardiovascular disease and diabetes. N. Engl. J. Med. 375, 2144–2153 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Mahajan, A. et al. Refining the accuracy of validated target identification through coding variant fine-mapping in type 2 diabetes. Preprint at https://www.biorxiv.org/content/early/2017/05/31/144410.1/ (2017).

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

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Grove, M.L. et al. Best practices and joint calling of the HumanExome BeadChip: the CHARGE Consortium. PLoS One 8, e68095 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tennessen, J.A. et al. Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science 337, 64–69 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 1000 Genomes Project Consortium. et al. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

  45. Zhan, X. & Liu, D.J. SEQMINER: an R-package to facilitate the functional interpretation of sequence-based associations. Genet. Epidemiol. 39, 619–623 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Fritsche, L.G. et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat. Genet. 48, 134–143 (2016).

    Article  CAS  PubMed  Google Scholar 

  47. Morris, A.P. et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat. Genet. 44, 981–990 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gaulton, K.J. et al. Genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci. Nat. Genet. 47, 1415–1425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. The UK Biobank. Genotyping and quality control of UK Biobank, a large-scale, extensively phenotyped prospective resource http://www.ukbiobank.ac.uk/wp-content/uploads/2014/04/UKBiobank_genotyping_QC_documentation-web.pdf (2015).

  51. 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 

  52. Baber, U. et al. Prevalence, impact, and predictive value of detecting subclinical coronary and carotid atherosclerosis in asymptomatic adults: the BioImage study. J. Am. Coll. Cardiol. 65, 1065–1074 (2015).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

D.J.L. is partially supported by R01HG008983 from the National Human Genome Research Institute of the National Institute of Health, and R21DA040177 and R01DA037904 from the National Institute of Drug Abuse of the National Institute of Health. G.M.P. is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health award K01HL125751. A.P.P. is supported by a research fellowship from the Stanley J. Sarnoff Cardiovascular Research Foundation. H. Tada is supported by a grant from the Japanese Circulation Society to study in the United States. The research was supported by the National Institute for Health Research (NIHR) Exeter Clinical Research Facility and ERC grant 323195; SZ-245 50371-GLUCOSEGENES-FP7-IDEAS-ERC to T.M.F. E.K.S. is supported by NIH grants R01 DK106621 and R01 DK107904, the University of Michigan Biological Sciences Scholars Program, and the University of Michigan Department of Internal Medicine. T.D.S. is supported by an ERC Advanced Principal Investigator award. A.P.M. is supported as a Wellcome Trust Senior Fellow in Basic Biomedical Science (grant no. WT098017). Y.E.C. is supported by HL117491 and HL129778 from the NIH. S.K.G. is supported by HL122684 from the NIH. P.L.A. is supported by NHLBI R21 HL121422-02 from the NIH. C.L., N.J.W., and R.A.S. acknowledge funding from the Medical Research Council, UK (MC_UU_12015/1). J.D. is supported as a British Heart Foundation Professor, European Research Council Senior Investigator, and National Institute for Health Research (NIHR) Senior Investigator. C.J.W. is supported by HL094535 and HL109946 from the NIH. S. Kathiresan is supported by a research scholar award from the Massachusetts General Hospital, the Donovan Family Foundation, and R01 HL127564 and R33 HL120781 from the NIH.

The views expressed in this manuscript are those of the authors and do not necessarily represent the views of the National Heart, Lung, and Blood Institute; the National Institutes of Health or the US Department of Health and Human Services.

This research has been conducted using the UK Biobank resource, application 7089. Funding support for participating studies in the meta-analysis can be found in the Supplementary Note.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

All authors contributed to and approved the results and comments on the manuscript. Writing: C.J.W., D.J.L., G.M.P., G.A., P.D., X.L., and S. Kathiresan. Study supervision: S. Kathiresan. Primary analysis: D.J.L. and G.M.P. Secondary analysis: A.V.K., A. Mahajan, Charge Diabetes Working Group, C.M.M., C.E., D.J.R., D.F.R., D.P., E.K.S., E.M.S., GOLD Consortium, J.B.M., J. Wessel, L.G.F., M.O.G., M.I.M., M. Boehnke, N. Stitziel, R.S.S., S. Somayajula, VA Million Veteran Program, and X.L. Functional characterization: A.R.T., C.A.C., H. Yu, K.M., N.W., and X.W. Contribution to study-specific analysis: A.S.B., A.C.A., A.C.M., A.D., A.-E.F., A.K.M., A. Langsted, A. Linneberg, A. Malarstig, A. Manichaikul, A. Maschio, A. Metspalu, A. Mulas, A.P., A.P.M., A.P.P., A.P.R., A.R., A.T.-H., A.U.J., A.V., A.V.S., A.Y.C., B.G.N., B.H.S., B.M.P., C.C., C.G., C.H., C.J.O'D., C.J.W., C.L., C.K., C.M.B., C.M.S., C.N.A.P., C.P., D. Alam, D. Arveiler, D.C.M.L., D.I.C., D.J.L., D.K., D.M.R., D.S., E.B., E.d.A., E.M., E.P.B., EPIC-CVD Consortium, The EPIC-InterAct Consortium, E.Z., F.B., F.C., F.G., F. Karpe, F. Kee, F.R., G.B.J., G. Davies, G. Dedoussis, G.E., G.M.P., G.P., H.A.K., H.G., H.M.S., H.R.W., H. Tada, H. Tang, H. Yaghootkar, H.Z., I.B., I.F., I.J.D., I.R., J.C.B., J.B.-J., J.C.C., J.C.D., J.D., J.D.R., J.F., J.G.W., J.H., J.I.R., J.J., J.K., J.M.C., J.M.M.H., J.M.J., J.M.O., J.M.S., J.B.N., J.N.H., J.S.K., J.-C.T., J.T., J.V., J. Weinstock, J.W.J., K.D.T., K.E.S., K.H., K.K., K.S., K.S.S., L.A.C., L.A.L., L.E.B., L.G., L.J.L., L.S., M. Benn, M. Brown, M.J.C., M.-P.D., M.E.G., M.E.J., M. Ferrario, M.F.F., M. Fornage, M.-R.J., M.J.N., M.L., M.L.G., M.M.-N., M.O.-M., M.P., M.W., M.X., M.Z., N.G., N.G.D.M., N.J.S., N.J.W., N.P., N.R.R., N.R.v.Z., N. Sattar, N.S.Z., O.L.H., O.M., O. Pedersen, O. Polasek, P.A., P.B.M., P.D., P.E.W., P.F., P.L.A., P. Mäntyselkä, P.M.R., P. Muntendam, P.R.K., P. Sever, P.S.T., P. Surendran, P.W.F., P.W.F.W., R.A.S., R.C., R.F.-S., R.J.F.L., R. Magi, R. Mehran, R.R., R.Y., S.P., S.F.N., S.J., S. Kanoni, S. Kathiresan, S.K.G., S.M.D., S. Sanna, S. Sivapalaratnam, S.S.R., S.T., T.B.H., T.D.S., T. Ebeling, T.E.-I.C., T. Esko, T.H., T.L.A., T. Lakka, T. Lauritzen, T.M.F., T.V.V., U.B., V.F., V.G., V.S., W.G., W. Zhang, W. Zhou, X.S., Y.E.C., Y.H., Y.-D.I.C., Y.L., Y. Zhang, and Y. Zhou.

Corresponding authors

Correspondence to Cristen J Willer or Sekar Kathiresan.

Ethics declarations

Competing interests

S. Kathiresan has received grant support from Bayer Healthcare and Amarin; holds equity in San Therapeutics and Catabasis; and has received personal fees for participation in scientific advisory boards for Bayer Healthcare, Catabasis, Regeneron Genetics Center, Merck, Celera, Genomics PLC, Novartis, Sanofi, AstraZeneca, Alnylam, Eli Lilly Company, Leerink Partners, Noble Insights, and Ionis Pharmaceuticals. All other authors have no relationships relevant to the contents of this paper to disclose. Merck authors are employees of Merck Sharp Dohme Corp., New Jersey, USA.

Additional information

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

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

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

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

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

Integrated supplementary information

Supplementary Figure 1 Quantile–quantile (QQ) plots of single-variant association analysis P values for each lipid trait.

Supplementary Figure 2 Manhattan plot of single-variant association analysis P values for LDL-C.

The novel coding variants are labeled in the plot.

Supplementary Figure 3 Manhattan plot of single-variant association analysis P values for HDL-C.

The novel coding variants are labeled in the plot.

Supplementary Figure 4 Manhattan plot of single-variant association analysis P values for triglycerides.

The novel coding variants are labeled in the plot.

Supplementary Figure 5 Manhattan plot of single-variant association analysis P values for total cholesterol.

The novel coding variants are labeled in the plot.

Supplementary Figure 6 Diagram of sequential forward-selection procedure.

Supplementary Figure 7 FPLC profile showing cholesterol content of plasma-lipoprotein fractions from pooled plasma of WT→Ldlr−/− or JAK2V617F→Ldlr−/− recipient mice fed a Western diet for 8 weeks.

Supplementary Figure 8 mRNA and protein levels of recombinantly expressed wild-type A1CF and p.Gly398Ser variant.

(a) mRNA levels of recombinantly expressed wild-type A1CF and p.Gly398Ser variant are similar in both Huh7 wild-type and A1CF knockout cells (labeled as A1CF KO). (b) and (c) APOB100 ELISA (b) and Western Blot (c) measuring cellular APOB100 levels in Huh7 wild-type and A1CF knockout cells. The bars of mean value and error bars of SD are showed in (a) and (b) from experiments with replicates, N=3 for a, N=4 for b. Statistically significant differences are marked (*p<0.05, **p<0.01).

Supplementary Figure 9 Knock-in mice for the A1CF p.Gly398Ser mutation.

(A) The sequences of the wild-type (WT) and A1cf p.Gly398Ser (KI) alleles. For the WT allele, the guide RNA protospacer is underlined, the PAM is in bold, and the base specifically affected by the A1cf p.Gly398Ser mutation is in red. For the KI allele, the altered bases are underlined, and the base specifically affected by the A1cf p.Gly398Ser mutation is in red. (B) The electropherogram is from a mouse heterozygous for the WT and KI alleles. The base specifically affected by the A1cf p.Gly398Ser mutation is shaded. (C) Plasma triglyceride levels of colony mates of the C57BL/6J background (N = 9 wild-type mice and 8 homozygous knock-in mice). Data are displayed as means and s.e.m. The P-value was calculated with the Mann-Whitney U test. (D) FPLC lipoprotein profiles for triglycerides or cholesterol in pooled plasma samples from WT and KI mice.

Supplementary Figure 10 Association of loss-of-function variants in HBB with hematologic traits and blood lipids.

Hematologic estimates are from an exome chip analysis of 24,814 individuals*. Lipid estimates are derived from fixed effects meta-analysis of estimates from the Global Lipids Genetics Consortium and Myocardial Infarction Genetics Consortium.

* Auer, P.L. et al. Rare and low-frequency coding variants in CXCR2 and other genes are associated with hematological traits. Nat Genet 46, 629-34 (2014).

Supplementary Figure 11 Association of HBB loss-of-function variants with coronary artery disease.

The association of loss of function variants with coronary artery disease was estimated using logistic regression with adjustment for age, sex and principal components of ancestry.

Supplementary Figure 12 Correlation plot of the effects of HDL and AMD for 168 independent HDL variants.

Supplementary Figure 13 Association of PCSK9 p.R46L with risk for type 2 diabetes.

In each study, the relationship of PCSK9 p.R46L with risk of type 2 diabetes was obtained. P-values for association tests and confidence intervals were determined using exact methods. A meta-analysis across studies was performed with the use of the Cochran–Mantel–Haenszel statistics for stratified 2-by-2 tables. This method combines score statistics and is particularly useful when some observed odds ratios are zero.

Supplementary Figure 14 Correlation plot of the effects of 113 independent LDL variants with MAF >1% and T2D.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14 and Supplementary Note

Life Sciences Reporting Summary

Supplementary Table 1

Studies contributing to meta-analysis

Supplementary Table 2

Descriptive statistics for lipid levels across contributing studies.

Supplementary Table 3

Genotyping and analysis methods across contributing studies

Supplementary Table 4

Variant site distribution by alternative allele frequency and annotations

Supplementary Table 5

Forty new loci where non-protein-altering variants are associated with lipid levels

Supplementary Table 6

Reason for non-coding variants on array

Supplementary Table 7

Association results in current study for 175 previously reported GWAS variants

Supplementary Table 8

Association analysis of novel lipid loci in samples of European American, African American, South Asian, and Hispanic ancestries.

Supplementary Table 9

Gene-level association results

Supplementary Table 10

Replication results for 75 novel primary associations

Supplementary Table 11

Variance explained by known and independently associated SNPs

Supplementary Table 12

Association Results for 444 independently associated variants with lipid traits

Supplementary Table 13

Loci where protein-altering variant is top signal or protein-altering variant explains the GWAS signal

Supplementary Table 14

59 loci where there's a protein-altering variant that is either the top signal, explains the signal or is independent.

Supplementary Table 15

Association results for null mutations with p < 0.001

Supplementary Table 16

HDL-C variants and risk for age-related macular degeneration (AMD)

Supplementary Table 17

DNA variants in CETP robustly associate with HDL-C and risk for AMD

Supplementary Table 18

Thirty studies from populations of European ancestry contributing to PCSK9 p.R46L on risk of T2D

Supplementary Table 19

Association of LDL-C variants with coronary artery disease (CAD) and type 2 diabetes (T2D)

Supplementary Table 20

Definitions of outcomes in UK Biobank PheWAS

Supplementary Table 21

sgRNA sequences for functional follow-up experiments

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, D., Peloso, G., Yu, H. et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nat Genet 49, 1758–1766 (2017). https://doi.org/10.1038/ng.3977

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3977

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