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.

  • Review Article
  • Published:

Multi-omics study for interpretation of genome-wide association study

Abstract

Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with complex traits, including a wide variety of diseases. Despite the successful identification of associated loci, interpreting GWAS findings remains challenging and requires other biological resources. Omics, including genomics, transcriptomics, proteomics, metabolomics, and epigenomics, are increasingly used in a broad range of research fields. Integrative analyses applying GWAS with these omics data are expected to expand our knowledge of complex traits and provide insight into the pathogenesis of complex diseases and their causative factors. Recently, associations between genetic variants and omics data have been comprehensively evaluated, providing new information on the influence of genetic variants on omics. Furthermore, recent advances in analytic methods, including single-cell technologies, have revealed previously unknown disease mechanisms. To advance our understanding of complex traits, integrative analysis using GWAS with multi-omics data is needed. In this review, I describe successful examples of integrative analyses based on omics and GWAS, discuss the limitations of current multi-omics analyses, and provide a perspective on future integrative studies.

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

Similar content being viewed by others

References

  1. Manolio TA. Genomewide association studies and assessment of the risk of disease. N Engl J Med. 2010;363:166–76.

    Article  CAS  PubMed  Google Scholar 

  2. Ozaki K, Ohnishi Y, Iida A, Sekine A, Yamada R, Tsunoda T, et al. Functional SNPs in the lymphotoxin-α gene that are associated with susceptibility to myocardial infarction. Nat Genet. 2002;32:650–4.

    Article  CAS  PubMed  Google Scholar 

  3. Shendure J, Findlay GM, Snyder MW. Genomic medicine–progress, pitfalls, and promise. Cell. 2019;177:45–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lee JJ, Wedow R, Okbay A, Kong E, Maghzian O, Zacher M, et al. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat Genet. 2018;50:1112–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Karlsson Linnér R, Biroli P, Kong E, Meddens SFW, Wedow R, Fontana MA, et al. Genome-wide association analyses of risk tolerance and risky behaviors in over 1 million individuals identify hundreds of loci and shared genetic influences. Nat Genet. 2019;51:245–57.

    Article  PubMed  CAS  Google Scholar 

  6. Yang J, Manolio TA, Pasquale LR, Boerwinkle E, Caporaso N, Cunningham JM, et al. Genome partitioning of genetic variation for complex traits using common SNPs. Nat Genet. 2011;43:519–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yang J, Bakshi A, Zhu Z, Hemani G, Vinkhuyzen AAE, Lee SH, et al. Genetic variance estimation with imputed variants finds negligible missing heritability for human height and body mass index. Nat Genet. 2015;47:1114–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, et al. Finding the missing heritability of complex diseases. Nature. 2009;461:747–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wuttke M, Li Y, Li M, Sieber KB, Feitosa MF, Gorski M, et al. A catalog of genetic loci associated with kidney function from analyses of a million individuals. Nat Genet. 2019;51:957–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Karczewski KJ, Snyder MP. Integrative omics for health and disease. Nat Rev Genet. 2018;19:299–310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nicolae DL, Gamazon E, Zhang W, Duan S, Eileen Dolan M, Cox NJ. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 2010;6:e1000888.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Takata A, Matsumoto N, Kato T. Genome-wide identification of splicing QTLs in the human brain and their enrichment among schizophrenia-associated loci. Nat Commun. 2017;8:14519.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Claussnitzer M, Dankel SN, Kim K-H, Quon G, Meuleman W, Haugen C, et al. FTO obesity variant circuitry and adipocyte browning in humans. N Engl J Med. 2015;373:895–907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Momozawa Y, Mni M, Nakamura K, Coppieters W, Almer S, Amininejad L, et al. Resequencing of positional candidates identifies low frequency IL23R coding variants protecting against inflammatory bowel disease. Nat Genet. 2011;43:43–7.

    Article  CAS  PubMed  Google Scholar 

  15. Rivas MA, Beaudoin M, Gardet A, Stevens C, Sharma Y, Zhang CK, et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat Genet. 2011;43:1066–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Fritsche LG, Igl W, Bailey JNC, Grassmann F, Sengupta S, Bragg-Gresham JL, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet. 2015;48:134–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Momozawa Y, Akiyama M, Kamatani Y, Arakawa S, Yasuda M, Yoshida S, 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. 2016;25:5027–34.

    CAS  PubMed  Google Scholar 

  18. Nica AC, Montgomery SB, Dimas AS, Stranger BE, Beazley C, Barroso I, et al. Candidate causal regulatory effects by integration of expression QTLs with complex trait genetic associations. PLoS Genet. 2010;6:e1000895.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Giambartolomei C, Vukcevic D, Schadt EE, Franke L, Hingorani AD, Wallace C, et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 2014;10:e1004383.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Chun S, Casparino A, Patsopoulos NA, Croteau-Chonka DC, Raby BA, et al. Limited statistical evidence for shared genetic effects of eQTLs and autoimmune-disease-associated loci in three major immune-cell types. Nat Genet. 2017;49:600–605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Akiyama M, Okada Y, Kanai M, Takahashi A, Momozawa Y, Ikeda M, et al. Genome-wide association study identifies 112 new loci for body mass index in the Japanese population. Nat Genet. 2017;49:1458–67.

    Article  CAS  PubMed  Google Scholar 

  22. Suzuki K, Akiyama M, Ishigaki K, Kanai M, Hosoe J, Shojima N, et al. Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet. 2019;51:379–86.

    Article  CAS  PubMed  Google Scholar 

  23. Gamazon ER, Wheeler HE, Shah KP, Mozaffari SV, Aquino-Michaels K, Carroll RJ, et al. A gene-based association method for mapping traits using reference transcriptome data. Nat Genet. 2015;47:1091–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gusev A, Ko A, Shi H, Bhatia G, Chung W, Penninx BWJH, et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat Genet. 2016;48:245–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wainberg M, Sinnott-Armstrong N, Mancuso N, Barbeira AN, Knowles DA, Golan D, et al. Opportunities and challenges for transcriptome-wide association studies. Nat Genet. 2019;51:592–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Westra H-J, Peters MJ, Esko T, Yaghootkar H, Schurmann C, Kettunen J, et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet. 2013;45:1238–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ratnapriya R, Sosina OA, Starostik MR, Kwicklis M, Kapphahn RJ, Fritsche LG, et al. Retinal transcriptome and eQTL analyses identify genes associated with age-related macular degeneration. Nat Genet. 2019;51:606–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ishigaki K, Kochi Y, Suzuki A, Tsuchida Y, Tsuchiya H, Sumitomo S, et al. Polygenic burdens on cell-specific pathways underlie the risk of rheumatoid arthritis. Nat Genet. 2017;49:1120–5.

    Article  CAS  PubMed  Google Scholar 

  29. Van Der Wijst MGP, Brugge H, De Vries DH, Deelen P, Swertz MA, Franke L. Single-cell RNA sequencing identifies celltype-specific cis-eQTLs and co-expression QTLs. Nat Genet. 2018;50:493–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. van der Wijst MGP, de Vries DH, Groot HE, Trynka G, Hon CC, Bonder MJ, et al. The single-cell eQTLGen consortium. Elife. 2020;9:e52155.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Menon M, Mohammadi S, Davila-Velderrain J, Goods BA, Cadwell TD, Xing Y, et al. Single-cell transcriptomic atlas of the human retina identifies cell types associated with age-related macular degeneration. Nat Commun. 2019;10:4902.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Orozco LD, Chen H, Cox C, Katschke KJ, Arceo R, Espiritu C, et al. Integration of eQTL and a single-cell atlas in the human eye identifies causal genes for age-related macular degeneration. Cell Rep. 2020;30:1246–1259.e6.

    Article  CAS  PubMed  Google Scholar 

  33. Sun BB, Maranville JC, Peters JE, Stacey D, Staley JR, Blackshaw J, et al. Genomic atlas of the human plasma proteome. Nature. 2018;558:73–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Suhre K, Arnold M, Bhagwat AM, Cotton RJ, Engelke R, Raffler J, et al. Connecting genetic risk to disease end points through the human blood plasma proteome. Nat Commun. 2017;8:14357.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Shin SY, Fauman EB, Petersen AK, Krumsiek J, Santos R, Huang J, et al. An atlas of genetic influences on human blood metabolites. Nat Genet. 2014;46:543–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Schlosser P, Li Y, Sekula P, Raffler J, Grundner-Culemann F, Pietzner M, et al. Genetic studies of urinary metabolites illuminate mechanisms of detoxification and excretion in humans. Nat Genet. 2020;52:167–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Park PJ. ChIP-seq: advantages and challenges of a maturing technology. Nat Rev Genet. 2009;10:669–680.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Grubert F, Zaugg JB, Kasowski M, et al. Genetic control of chromatin states in humans involves local and distal chromosomal interactions. Cell. 2015;162:1051–1065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Tehranchi AK, Myrthil M, Martin T, Hie BL, Golan D, Fraser HB. Pooled ChIP-seq links variation in transcription factor binding to complex disease risk. Cell. 2016;165:730–741.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Reshef YA, Finucane HK, Kelley DR, et al. Detecting genome-wide directional effects of transcription factor binding on polygenic disease risk. Nat Genet. 2018;50:1483–1493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kumasaka N, Knights AJ, Gaffney DJ. Fine-mapping cellular QTLs with RASQUAL and ATAC-seq. Nat Genet. 2016;48:206–213.

    Article  CAS  PubMed  Google Scholar 

  42. Bulik-Sullivan BK, Loh P-R, Finucane HK, Ripke S, Yang J, Patterson N, et al. LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat Genet. 2015;47:291–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. The GTEx Consortium, Welter D, MacArthur J, Morales J, Burdett T, Hall P, et al. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348:648–60.

    Article  CAS  Google Scholar 

  44. Nielsen JB, Thorolfsdottir RB, Fritsche LG, Zhou W, Skov MW, Graham SE, et al. Biobank-driven genomic discovery yields new insight into atrial fibrillation biology. Nat Genet. 2018;50:1234–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pers TH, Karjalainen JM, Chan Y, Westra H-J, Wood AR, Yang J, et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat Commun. 2015;6:5890.

    Article  CAS  PubMed  Google Scholar 

  46. Trynka G, Sandor C, Han B, Xu H, Stranger BE, Liu XS, et al. Chromatin marks identify critical cell types for fine mapping complex trait variants. Nat Genet. 2013;45:124–30.

    Article  CAS  PubMed  Google Scholar 

  47. Finucane HK, Bulik-Sullivan B, Gusev A, Trynka G, Reshef Y, Loh P-R, et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat Genet. 2015;47:1228–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Finucane HK, Reshef YA, Anttila V, Slowikowski K, Gusev A, Byrnes A, et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat Genet. 2018;50:621–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sakaue S, Hirata J, Maeda Y, Kawakami E, Nii T, Kishikawa T, et al. Integration of genetics and miRNA–target gene network identified disease biology implicated in tissue specificity. Nucleic Acids Res. 2018;46:11898–909.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hon CC, Ramilowski JA, Harshbarger J, Bertin N, Rackham OJL, Gough J, et al. An atlas of human long non-coding RNAs with accurate 5′ ends. Nature. 2017;543:199–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Liu J, van Klinken JB, Semiz S, van Dijk KW, Verhoeven A, Hankemeier T, et al. A mendelian randomization study of metabolite profiles, fasting glucose, and type 2 diabetes. Diabetes. 2017;66:2915–26.

    Article  CAS  PubMed  Google Scholar 

  52. Schaid DJ, Chen W, Larson NB. From genome-wide associations to candidate causal variants by statistical fine-mapping. Nat Rev Genet. 2018;19:491–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Solovieff N, Cotsapas C, Lee PH, Purcell SM, Smoller JW. Pleiotropy in complex traits: challenges and strategies. Nat Rev Genet. 2013;14:483–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kishikawa T, Maeda Y, Nii T, Motooka D, Matsumoto Y, Matsushita M, et al. Metagenome-wide association study of gut microbiome revealed novel aetiology of rheumatoid arthritis in the Japanese population. Ann Rheum Dis. 2020;79:103–11.

    Article  CAS  PubMed  Google Scholar 

  55. Oki S, Ohta T, Shioi G, Hatanaka H, Ogasawara O, Okuda Y, et al. Ch IP ‐Atlas: a data‐mining suite powered by full integration of public Ch IP ‐seq data. EMBO Rep. 2018;19:1–10.

    Article  CAS  Google Scholar 

  56. Meehan TF, Conte N, West DB, Jacobsen JO, Mason J, Warren J, et al. Disease model discovery from 3,328 gene knockouts by The International Mouse Phenotyping Consortium. Nat Genet. 2017;49:1231–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46:D1074–82.

    Article  CAS  PubMed  Google Scholar 

  58. Li YH, Yu CY, Li XX, Zhang P, Tang J, Yang Q, et al. Therapeutic target database update 2018: enriched resource for facilitating bench-to-clinic research of targeted therapeutics. Nucleic Acids Res. 2018;46:D1121–7.

    Article  CAS  PubMed  Google Scholar 

  59. Torkamani A, Wineinger NE, Topol EJ. The personal and clinical utility of polygenic risk scores. Nat Rev Genet. 2018;19:581–590.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Masato Akiyama.

Ethics declarations

Conflict of interest

MA is the endowed chair faculty member supported by NIDEK Company Ltd.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Akiyama, M. Multi-omics study for interpretation of genome-wide association study. J Hum Genet 66, 3–10 (2021). https://doi.org/10.1038/s10038-020-00842-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s10038-020-00842-5

This article is cited by

Search

Quick links