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:

Exploring the genetic architecture of inflammatory bowel disease by whole-genome sequencing identifies association at ADCY7

Abstract

To further resolve the genetic architecture of the inflammatory bowel diseases ulcerative colitis and Crohn's disease, we sequenced the whole genomes of 4,280 patients at low coverage and compared them to 3,652 previously sequenced population controls across 73.5 million variants. We then imputed from these sequences into new and existing genome-wide association study cohorts and tested for association at 12 million variants in a total of 16,432 cases and 18,843 controls. We discovered a 0.6% frequency missense variant in ADCY7 that doubles the risk of ulcerative colitis. Despite good statistical power, we did not identify any other new low-frequency risk variants and found that such variants explained little heritability. We detected a burden of very rare, damaging missense variants in known Crohn's disease risk genes, suggesting that more comprehensive sequencing studies will continue to improve understanding of the biology of complex diseases.

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: Overview of our study.
Figure 2: Association analysis for the NOD2ADCY7 region on chromosome 16.
Figure 3: Associations between NOD2 variants and Crohn's disease.
Figure 4: Burden of rare, damaging variants in Crohn's disease.
Figure 5: Relative power of this study in comparison to previous GWAS.

Similar content being viewed by others

References

  1. Liu, J.Z. et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 47, 979–986 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Parkes, M. et al. Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn's disease susceptibility. Nat. Genet. 39, 830–832 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yamazaki, K. et al. A genome-wide association study identifies 2 susceptibility loci for Crohn's disease in a Japanese population. Gastroenterology 144, 781–788 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Anderson, C.A. et al. Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat. Genet. 43, 246–252 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kenny, E.E. et al. A genome-wide scan of Ashkenazi Jewish Crohn's disease suggests novel susceptibility loci. PLoS Genet. 8, e1002559 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Julià, A. et al. A genome-wide association study identifies a novel locus at 6q22.1 associated with ulcerative colitis. Hum. Mol. Genet. 23, 6927–6934 (2014).

    Article  PubMed  CAS  Google Scholar 

  7. Yang, S.-K. et al. Genome-wide association study of Crohn's disease in Koreans revealed three new susceptibility loci and common attributes of genetic susceptibility across ethnic populations. Gut 63, 80–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Ellinghaus, D. et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat. Genet. 48, 510–518 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Manolio, T.A. et al. Finding the missing heritability of complex diseases. Nature 461, 747–753 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zuk, O. et al. Searching for missing heritability: designing rare variant association studies. Proc. Natl. Acad. Sci. USA 111, E455–E464 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rivas, M.A. et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat. Genet. 43, 1066–1073 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Beaudoin, M. et al. Deep resequencing of GWAS loci identifies rare variants in CARD9, IL23R and RNF186 that are associated with ulcerative colitis. PLoS Genet. 9, e1003723 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hunt, K.A. et al. Negligible impact of rare autoimmune-locus coding-region variants on missing heritability. Nature 498, 232–235 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Prescott, N.J. et al. Pooled sequencing of 531 genes in inflammatory bowel disease identifies an associated rare variant in BTNL2 and implicates other immune related genes. PLoS Genet. 11, e1004955 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Do, R. et al. Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature 518, 102–106 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. De Rubeis, S. et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 515, 209–215 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Singh, T. et al. Rare loss-of-function variants in SETD1A are associated with schizophrenia and developmental disorders. Nat. Neurosci. 19, 571–577 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Huang, H. et al. Association mapping of inflammatory bowel disease loci to single variant resolution. Preprint at bioRxiv http://dx.doi.org/10.1101/028688 (2015).

  19. Farh, K.K.-H. et al. Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518, 337–343 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Li, Y., Sidore, C., Kang, H.M., Boehnke, M. & Abecasis, G.R. Low-coverage sequencing: implications for design of complex trait association studies. Genome Res. 21, 940–951 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. CONVERGE Consortium. Sparse whole-genome sequencing identifies two loci for major depressive disorder. Nature 523, 588–591 (2015).

  22. Danjou, F. et al. Genome-wide association analyses based on whole-genome sequencing in Sardinia provide insights into regulation of hemoglobin levels. Nat. Genet. 47, 1264–1271 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. UK10K Consortium. The UK10K project identifies rare variants in health and disease. Nature 526, 82–90 (2015).

  24. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Browning, B.L. & Browning, S.R. Improving the accuracy and efficiency of identity-by-descent detection in population data. Genetics 194, 459–471 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Handsaker, R.E. et al. Large multiallelic copy number variations in humans. Nat. Genet. 47, 296–303 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007).

  30. Barrett, J.C. et al. Genome-wide association study of ulcerative colitis identifies three new susceptibility loci, including the HNF4A region. Nat. Genet. 41, 1330–1334 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Durbin, R. Efficient haplotype matching and storage using the positional Burrows–Wheeler transform (PBWT). Bioinformatics 30, 1266–1272 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. de Lange, K.M. et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat. Genet. http://dx.doi.org/10.1038/ng.3760 (2017).

  33. Li, Y.R. et al. Meta-analysis of shared genetic architecture across ten pediatric autoimmune diseases. Nat. Med. 21, 1018–1027 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dahle, M.K., Myhre, A.E., Aasen, A.O. & Wang, J.E. Effects of forskolin on Kupffer cell production of interleukin-10 and tumor necrosis factor α differ from those of endogenous adenylyl cyclase activators: possible role for adenylyl cyclase 9. Infect. Immun. 73, 7290–7296 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Duan, B. et al. Distinct roles of adenylyl cyclase VII in regulating the immune responses in mice. J. Immunol. 185, 335–344 (2010).

    Article  CAS  PubMed  Google Scholar 

  36. Jiang, L.I., Sternweis, P.C. & Wang, J.E. Zymosan activates protein kinase A via adenylyl cyclase VII to modulate innate immune responses during inflammation. Mol. Immunol. 54, 14–22 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Risøe, P.K. et al. Higher TNFα responses in young males compared to females are associated with attenuation of monocyte adenylyl cyclase expression. Hum. Immunol. 76, 427–430 (2015).

    Article  PubMed  CAS  Google Scholar 

  38. Pierre, S., Eschenhagen, T., Geisslinger, G. & Scholich, K. Capturing adenylyl cyclases as potential drug targets. Nat. Rev. Drug Discov. 8, 321–335 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Bhatia, G. et al. Subtle stratification confounds estimates of heritability from rare variants. Preprint at bioRxiv http://dx.doi.org/10.1101/048181 (2016).

  40. Yang, J., Lee, S.H., Goddard, M.E. & Visscher, P.M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chen, G.-B. et al. Estimation and partitioning of (co)heritability of inflammatory bowel disease from GWAS and Immunochip data. Hum. Mol. Genet. 23, 4710–4720 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Purcell, S.M. et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature 506, 185–190 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Derkach, A. et al. Association analysis using next-generation sequence data from publicly available control groups: the robust variance score statistic. Bioinformatics 30, 2179–2188 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Andersson, R. et al. An atlas of active enhancers across human cell types and tissues. Nature 507, 455–461 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zimmerman, N.P., Kumar, S.N., Turner, J.R. & Dwinell, M.B. Cyclic AMP dysregulates intestinal epithelial cell restitution through PKA and RhoA. Inflamm. Bowel Dis. 18, 1081–1091 (2012).

    Article  PubMed  Google Scholar 

  46. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Genovese, G. et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat. Neurosci. 19, 1433–1441 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Jostins, L. et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hinrichs, A.S. et al. The UCSC Genome Browser Database: update 2006. Nucleic Acids Res. 34, D590–D598 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. 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 

  51. Marchini, J. & Howie, B. Genotype imputation for genome-wide association studies. Nat. Rev. Genet. 11, 499–511 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Speed, D. & Balding, D.J. MultiBLUP: improved SNP-based prediction for complex traits. Genome Res. 24, 1550–1557 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all individuals who contributed samples to the study. This work was co-funded by the Wellcome Trust (098051) and the Medical Research Council, UK (MR/J00314X/1). Case collections were supported by Crohn's and Colitis UK. K.M.d.L., L.M., Y.L., C.A.L., C.A.A. and J.C.B. are supported by the Wellcome Trust (098051; 093885/Z/10/Z). K.M.d.L. is supported by a Woolf Fisher Trust scholarship. C.A.L. is a clinical lecturer funded by the NIHR. H.U. is supported by the Crohn's and Colitis Foundation of America (CCFA) and the Leona M. and Harry B. Helmsley Charitable Trust. We acknowledge support from the UK Department of Health via NIHR comprehensive Biomedical Research Centre awards to Guy's and St Thomas' NHS Foundation Trust in partnership with King's College London and to Addenbrooke's Hospital, Cambridge, in partnership with the University of Cambridge, and the BRC to the Oxford IBD cohort study, University of Oxford. This research was also supported by the NIHR Newcastle Biomedical Research Centre. The UK Household Longitudinal Study is led by the Institute for Social and Economic Research at the University of Essex and funded by the Economic and Social Research Council. The survey was conducted by NatCen, and the genome-wide scan data were analyzed and deposited by the Wellcome Trust Sanger Institute. Information on how to access the data can be found on the Understanding Society website. We are grateful for genotyping data from the British Society for Surgery of the Hand Genetics of Dupuytren's Disease consortium and L. Southam for assistance with genotype intensities. This research has been conducted using the UK Biobank Resource.

Author information

Authors and Affiliations

Authors

Contributions

Y.L., K.M.d.L., L.J., L.M., J.C.B. and C.A.A. performed statistical analysis. Y.L., K.M.d.L., L.J., L.M., J.C.L., C.A.L., E.G.S., J.R., M. Pollard, S.N. and S.M. processed the data. T.A., C.E., N.A.K., A.H., C.H., J.C.M., J.C.L., C.M., W.G.N., J.S., A.S., M.T., H.U., D.C.W., N.J.P., C.W.L., M. Parkes and C.G.M. contributed samples and/or materials. Y.L., K.M.d.L., L.M., J.C.L., M. Parkes, C.A.L., N.A.K., J.C.B. and C.A.A. wrote the manuscript. All authors read and approved the final version of the manuscript. J.C.M., M. Parkes, C.W.L., T.A., N.J.P., J.C.B. and C.A.A. conceived and designed experiments.

Corresponding authors

Correspondence to Jeffrey C Barrett or Carl A Anderson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Production pipeline of SNVs and indels.

Supplementary Figure 2 Genotypic accuracy of sequencing data.

Dosage r2 plots for determining sequencing quality when compared against other genotyping data. The x axis is minor allele frequency calculated based on sequencing samples, and the y axis is correlation between dosages for sequencing and genotype data sets. Numbers in parentheses are the number of individuals with both types of data.

Supplementary Figure 3 Biallelic SNV discovery rate compared to the 1000 Genomes Project Phase 3 European panel.

Percentage of biallelic SNVs in all autosomal regions that are shared by the IBD sequencing set and 1000 Genomes Project (1000GP) Phase 3 European panel (503 individuals). Left, percentage of IBD sequencing SNVs that are also found in 1000GP; right, variants identified in the 1000GP set that are also in the IBD sequencing cohort. MAFs on the left were calculated based on SNVs discovered in the IBD sequencing project, and MAFs on the right were calculated based on the 1000GP set. Different lines represent SNVs in different quality control stages of the analysis.

Supplementary Figure 4 Number of copy number variants called per cohort.

Average number of calls per individual per site, across different copy number variant (CNV) lengths. UK10K controls (6×) are shown in yellow, Crohn’s disease cases (4×) are shown in red, and ulcerative colitis cases (2×) are shown in blue.

Supplementary Figure 5 Quantile–quantile plots of genome-wide association studies for variants with MAF ≥ 0.1% in the sequencing data set.

λ1,000 values are reported for ulcerative colitis, Crohn’s disease and inflammatory bowel disease analyses. Gray shapes show the 95% confidence interval.

Supplementary Figure 6 Cluster plots for rs78534766.

(a–c) Cluster plots are shown for rs78534766 for the GWAS3 (a), replication (b) and UK Biobank (c) samples that passed quality control. SNP genotypes have been assigned based on cluster formation in scatterplots of normalized allele intensities X and Y. Each circle represents one individual’s genotype. Blue and red clouds correspond to homozygote genotypes for the SNP (CC/AA), green clouds correspond to the heterozygote genotype (CA) and gray clouds correspond to undetermined genotype.

Supplementary Figure 7 Workflow for heritability estimation.

Supplementary Figure 8 Distribution of INFO scores by cohort, across a range of minor allele frequencies.

(a) INFO scores calculated using genotype probabilities generated directly from the SAMtools Genotype Quality (GQ) field. (b) INFO scores calculated using genotype probabilities after imputation improvement using BEAGLE.

Supplementary Figure 9 Manhattan and quantile–quantile plots showing the results of gene-based burden tests using rare, functional coding variation.

Supplementary Figure 10 Principal-component analysis of the sequencing samples.

IBD samples are plotted with 11 different HapMap 3 populations.

Supplementary Figure 11 Effect of read depth on sensitivity and specificity across the allele frequency spectrum (UC (2×) CD (4×), controls (6×)).

(a–c) Top, full distribution of variant counts per individual at singletons (observed once in the data set) (a), doubletons (observed twice) (b) and variants with a MAF of 5% (c). (d) Plot showing the median of each distribution across a range of MAF values.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11, Supplementary Tables 1–17 and Supplementary Note (PDF 3457 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luo, Y., de Lange, K., Jostins, L. et al. Exploring the genetic architecture of inflammatory bowel disease by whole-genome sequencing identifies association at ADCY7. Nat Genet 49, 186–192 (2017). https://doi.org/10.1038/ng.3761

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3761

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