The genetic architecture of type 2 diabetes

Abstract

The genetic architecture of common traits, including the number, frequency, and effect sizes of inherited variants that contribute to individual risk, has been long debated. Genome-wide association studies have identified scores of common variants associated with type 2 diabetes, but in aggregate, these explain only a fraction of the heritability of this disease. Here, to test the hypothesis that lower-frequency variants explain much of the remainder, the GoT2D and T2D-GENES consortia performed whole-genome sequencing in 2,657 European individuals with and without diabetes, and exome sequencing in 12,940 individuals from five ancestry groups. To increase statistical power, we expanded the sample size via genotyping and imputation in a further 111,548 subjects. Variants associated with type 2 diabetes after sequencing were overwhelmingly common and most fell within regions previously identified by genome-wide association studies. Comprehensive enumeration of sequence variation is necessary to identify functional alleles that provide important clues to disease pathophysiology, but large-scale sequencing does not support the idea that lower-frequency variants have a major role in predisposition to type 2 diabetes.

Figures

Extended Data Figure 1. Summary of samples…
Extended Data Figure 1. Summary of samples and quality control procedures
This figure summarises data generation for whole genome sequencing (GoT2D), exome sequencing (GoT2D and T2D-GENES) and exome array genotyping (DIAGRAM). In addition, GoT2D whole genome sequence data was imputed into GWAS data from 44,414 subjects of European descent.
Extended Data Figure 4. Power for single…
Extended Data Figure 4. Power for single and aggregate variant association
a-g. Power to detect single-variant association (α=5×10−8) at varying minor allele frequency (x-axis) and allelic odds-ratio (y-axis) for seven effective sample size (Neff) scenarios relevant to the genomes (a-c) and exomes (dg) component of this project. a. variant observed in 2,657 samples (the effective size of the GoT2D integrated panel); b. variant observed in 28,350 samples (the effective size of the imputed data set); c. variant observed in the GoT2D integrated panel and the imputed data set (effective sample size 31,007);d. ancestry-specific variant in 2,000 samples (the size of each of the non-European exome sequence data sets); e. European specific variant in 5,000 samples (the combined size of the European exome sequence data sets);f. variant observed with shared frequency across all ancestry groups in 12,940 samples (the size of the combined exome sequence data set); and g.variant observed in the combined exome array and sequencing data set (effective sample size 82,758). h-i. Power for gene based test of association (SKAT-O) according to liability variance explained. In h, 50% of the variants contribute to disease risk while the remaining 50% have no effect on disease risk; ini., 100% of the variants contribute to disease risk. For each, sample sizes considered are 2,000 (ancestry-specific effects; green) and 12,940 (ancestry-shared effects; blue). Power is shown for two levels of significance (α=2.5×10−6 and α=0.001). From these simulation studies, it is clear that under the optimistic model, where effects are shared across all ethnicities (blue line) and all variants contribute, power is >60% for 1% variance explained and α=2.5×10−6. However, power declines rapidly if either criterion is relaxed.
Extended Data Figure 6. Single variant analyses
Extended Data Figure 6. Single variant analyses
Manhattan plot of single-variant analyses generated from a.exome sequence data in 6,504 cases and 6,436 controls of African American, East Asian, European, Hispanic, and South Asian ancestry; b. exome array genotypes in 28,305 cases and 51,549 controls of European ancestry; and c. combined meta-analysis of exome array and exome sequence samples. Coding variants are categorized according to their relationships to the previously reported lead variant from GWAS region. Loci achieving genome-wide significance only in the combined analysis are highlighted in bold. The HNF1A variant reaching genome-wide significance in the combined analysis is a synonymous variant (Thr515Thr). The dashed horizontal line in each panel designates the threshold for genome-wide significance (p<5×10−8).
Extended Data Figure 7. Classification of coding…
Extended Data Figure 7. Classification of coding variants according to their relationship to reported lead variants for each GWAS region
The ideogram shows the location of 25 coding variant associations at 16 loci described in the text. The number in each circle corresponds to the number of associated variants at each locus. Variants are grouped into five categories based on inferred relationship with the GWAS lead variant. For some of these categories, the figure includes representative regional association plots based on exome array meta-analysis data from 28,305 cases and 51,549 controls. The locus displayed for each category is designated in bold. The first plot in each panel shows the unconditional association results; middle plot the association results after conditioning on the non-coding GWAS SNP; and the last plot the results after conditioning on the most significantly associated coding variant. Each point represents a SNP in the exome array meta-analysis, plotted with their p-value (on a –log10 scale) as a function of the genomic position (hg19). In each panel, the lead coding variant is represented by the purple symbol. The color-coding of all other SNPs indicates LD with the lead SNP (estimated by European r2 from 1000 Genomes March 2012 reference panel: red r2≥0.8; gold 0.6≤r2<0.8; green 0.4≤r2<0.6; cyan 0.2≤r2<0.4; blue r2<0.2; grey r2unknown). Gene annotations are taken from the University of California Santa Cruz genome browser. GWS: genome-wide significance. *Seven variants, three at ASCC2, and one each atTHADA, TSPAN8, FES andHNF4A did not achieve genome-wide significance themselves, but are included because they fall into genes and/or regions with other significant association signals (see text).
Extended Data Figure 9. Exclusion of synthetic…
Extended Data Figure 9. Exclusion of synthetic associations and construction of credible causal variant sets at T2D GWAS loci
Ten T2D GWAS loci were selected for synthetic association testing (p<0.001; Methods). a, The effect size observed at the GWAS index SNV (sequence data) before (navy blue) and after (light blue, grey) conditioning on candidate rare and low-frequency (MAF<5%) variants which could produce synthetic association. b, Example of synthetic association exclusion at the TCF7L2 locus. c, Credible sets for T2D GWAS loci where credible set consisted of <80 variants displaying the proportion of credible set variants present in the HapMap and 1000G catalogs.
Extended Data Figure 10
Extended Data Figure 10
Genome enrichment analysis in GoT2D whole genome sequence data (n=2,657) a, Functional annotation categories were defined using transcription, chromatin state and transcription factor binding data from GENCODE, ENCODE and other studies. b, T2D association statistics for variants at each T2D locus were jointly modelled with functional annotation using fgwas. In the resulting model we identified enrichment of coding exons (CDS), transcription factor binding sites (TFBS), mature adipose active enhancers and promoters (hASC-t4 EnhA, TssA), pancreatic islet active and weak enhancers (HI EnhA, EnhWk), pre-adipose active and weak enhancers (hASC-t1 EnhA, EnhWk), embryonic stem cell active promoters (H1-hESC TssA) and 5’ UTR. Dots represent enrichment estimates and horizontal lines the 95% confidence intervals. c, At the CCND2 locus, three variants not present in HapMap2 have a combined 90% posterior probability of being causal (rs4238013, rs3217801, rs73040004). One of these variants, rs3217801, is a 2-bp indel that overlaps an islet enhancer element.
Extended Data Figure 11. Low frequency variants…
Extended Data Figure 11. Low frequency variants in exome array data
Results from meta-analysis of 43,045 low-frequency and common coding variants on the exome array (assayed in 79,854 European subjects). a. Observed allelic ORs as a property of allele MAF. Variants missing in >8 cohorts or polymorphic in only one cohort were excluded. Colored lines represent contours for liability variance explained. Regions shaded grey denote ranges of OR and MAF consistent with 80% power (in this case, at α=5×10−7) to detect single-variant associations in this data set (given the observed range of missing data). Variants with a black collar are those highlighted by a bounding analysis as having a probability>0.8 of having LVE>0.1%; b. Distribution of each variant in the MAF/OR space was computed by assuming T2D prevalence of 8% and a beta and normal distribution for MAF and OR respectively. Probability is obtained by integrating the joint MAF-OR distributions over ranges of LVE; c. Single variant association, liability and bounding results for the known T2D GWAS variants on the exome array (Methods).
Figure 1. Ascertainment of variants and single-variant…
Figure 1. Ascertainment of variants and single-variant results
a, Sensitivity of low-coverage genome sequence data to detect SNVs in the deep exome sequence data, relative to other variant catalogs. Points represent results for a specific minor allele count. All results assume OR=1 for all variants, unless stated otherwise. Manhattan plots of single-variant association analyses for: b, sequence data alone (1,326 cases and 1,331 controls) and c, meta-analysis of sequence and imputed data (total of 14,297 cases and 32,774 controls).
Figure 2. Association between T2D and variants…
Figure 2. Association between T2D and variants in genes for Mendelian forms of diabetes
a, p-values of aggregate association for variants from 6,504 T2D cases and 6,436 controls in three sets of Mendelian diabetes genes, for five variant “masks” (Methods). Dotted line:p=0.05. b, Estimated T2D odds ratio (OR) for carriers of variants in each gene-set and mask. Error bars: one standard error. c, Estimated ORs (bars, left axis) and p-values (dots, right axis) for carriers of variants in the PTV+NSstrict mask for each gene. Error bars: one standard error. Red: OR > 1; blue: OR < 1; dotted line:p=0.05.
Figure 3. Empirical T2D association results compared…
Figure 3. Empirical T2D association results compared to results under different simulated disease models
Observed number of rare and low-frequency (MAFT and degree of coupling τ between the causal effects of variants and the selective pressure against them. Simulated data were generated to match GoT2D imputation quality as a function of MAF (Methods).

References

    1. Willemsen G, et al. The concordance and heritability of type 2 diabetes in 34,166 Twin Pairs From International Twin Registers: The Discordant Twin (DISCOTWIN) Consortium. Twin Res Hum Genet. 2015;18:762–71.
    1. Morris AP, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet. 2012;44:981–90.
    1. Mahajan A, et al. Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility. Nat Genet. 2014;46:234–44.
    1. Voight BF, et al. Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat Genet. 2010;42:579–89.
    1. Kooner JS, et al. Genome-wide association study in individuals of South Asian ancestry identifies six new type 2 diabetes susceptibility loci. Nat Genet. 2011;43:984–9.
    1. Cho YS, et al. Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians. Nat Genet. 2012;44:67–72.
    1. Steinthorsdottir V, et al. Identification of low-frequency and rare sequence variants associated with elevated or reduced risk of type 2 diabetes. Nat Genet. 2014;46:294–8.
    1. Ma RC, et al. Genome-wide association study in a Chinese population identifies a susceptibility locus for type 2 diabetes at 7q32 near PAX4. Diabetologia. 2013;56:1291–305.
    1. Huyghe JR, et al. Exome array analysis identifies new loci and low-frequency variants influencing insulin processing and secretion. Nat Genet. 2013;45:197–201.
    1. Gaulton KJ, et al. Genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci. Nat Genet. 2015;47:1415–25.
    1. Manolio TA, et al. Finding the missing heritability of complex diseases. Nature. 2009;461:747–53.
    1. Lohmueller KE, et al. Whole-exome sequencing of 2,000 Danish individuals and the role of rare coding variants in type 2 diabetes. Am J Hum Genet. 2013;93:1072–86.
    1. Albrechtsen A, et al. Exome sequencing-driven discovery of coding polymorphisms associated with common metabolic phenotypes. Diabetologia. 2013;56:298–310.
    1. Claussnitzer M, et al. Leveraging cross-species transcription factor binding site patterns: from diabetes risk loci to disease mechanisms. Cell. 2014;156:343–58.
    1. Lee S, Teslovich TM, Boehnke M, Lin X. General framework for meta-analysis of rare variants in sequencing association studies. Am J Hum Genet. 2013;93:42–53.
    1. Kang HM, et al. Variance component model to account for sample structure in genome-wide association studies. Nat Genet. 2010;42:348–54.
    1. Collombat P, et al. Opposing actions of Arx and Pax4 in endocrine pancreas development. Genes Dev. 2003;17:2591–603.
    1. Kooptiwut S, et al. Defective PAX4 R192H transcriptional repressor activities associated with maturity onset diabetes of the young and early onset-age of type 2 diabetes. J Diabetes Complications. 2012;26:343–7.
    1. InterAct Consortium et al. Design and cohort description of the InterAct Project: an examination of the interaction of genetic and lifestyle factors on the incidence of type 2 diabetes in the EPIC Study. Diabetologia. 2011;54:2272–82.
    1. Oppelt A, et al. Production of phosphatidylinositol 5-phosphate via PIKfyve and MTMR3 regulates cell migration. EMBO Rep. 2013;14:57–64.
    1. Kozlitina J, et al. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2014;46:352–6.
    1. Mahdessian H, et al. TM6SF2 is a regulator of liver fat metabolism influencing triglyceride secretion and hepatic lipid droplet content. Proc Natl Acad Sci U S A. 2014;111:8913–8.
    1. Thiagalingam A, Lengauer C, Baylin SB, Nelkin BD. RREB1, a ras responsive element binding protein, maps to human chromosome 6p25. Genomics. 1997;45:630–2.
    1. Murphy R, Ellard S, Hattersley AT. Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes. Nat Clin Pract Endocrinol Metab. 2008;4:200–13.
    1. Dickson SP, Wang K, Krantz I, Hakonarson H, Goldstein DB. Rare variants create synthetic genome-wide associations. PLoS Biol. 2010;8:e1000294.
    1. Anderson CA, Soranzo N, Zeggini E, Barrett JC. Synthetic associations are unlikely to account for many common disease genome-wide association signals. PLoS Biol. 2011;9:e1000580.
    1. Wray NR, Purcell SM, Visscher PM. Synthetic associations created by rare variants do not explain most GWAS results. PLoS Biol. 2011;9:e1000579.
    1. Sim X, et al. Transferability of type 2 diabetes implicated loci in multi-ethnic cohorts from Southeast Asia. PLoS Genet. 2011;7:e1001363.
    1. Goldstein DB. The importance of synthetic associations will only be resolved empirically. PLoS Biol. 2011;9:e1001008.
    1. Wakefield J. A Bayesian measure of the probability of false discovery in genetic epidemiology studies. Am J Hum Genet. 2007;81:208–27.
    1. Wellcome Trust Case Control Consortium et al. Bayesian refinement of association signals for 14 loci in 3 common diseases. Nat Genet. 2012;44:1294–301.
    1. Encode Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74.
    1. Mikkelsen TS, et al. Comparative epigenomic analysis of murine and human adipogenesis. Cell. 2010;143:156–69.
    1. Parker SC, et al. Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants. Proc Natl Acad Sci U S A. 2013;110:17921–6.
    1. Pasquali L, et al. Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nat Genet. 2014;46:136–43.
    1. Gaulton KJ, et al. A map of open chromatin in human pancreatic islets. Nat Genet. 2010;42:255–9.
    1. Maurano MT, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science. 2012;337:1190–5.
    1. Pickrell JK. Joint analysis of functional genomic data and genome-wide association studies of 18 human traits. Am J Hum Genet. 2014;94:559–73.
    1. Falconer DS. The inheritance of liability to certain diseases, estimated from the incidence among relatives. Ann Hum Genet. 1965;29:51–76.
    1. Agarwala V, Flannick J, Sunyaev S. GoT2D Consortium & Altshuler, D. Evaluating empirical bounds on complex disease genetic architecture. Nat Genet. 2013;45:1418–27.
    1. McClellan J, King MC. Genetic heterogeneity in human disease. Cell. 2010;141:210–7.
    1. Yang J, et al. Common SNPs explain a large proportion of the heritability for human height. Nat Genet. 2010;42:565–9.
    1. Flannick J, et al. Loss-of-function mutations in SLC30A8 protect against type 2 diabetes. Nat Genet. 2014;46:357–63.
    1. Bonnefond A, et al. Rare MTNR1B variants impairing melatonin receptor 1B function contribute to type 2 diabetes. Nat Genet. 2012;44:297–301.
    1. Sigma Type 2 Diabetes Consortium et al. Sequence variants in SLC16A11 are a common risk factor for type 2 diabetes in Mexico. Nature. 2014;506:97–101.
    1. Moltke I, et al. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Nature. 2014;512:190–3.
    1. Sigma Type 2 Diabetes Consortium et al. Association of a low-frequency variant in HNF1A with type 2 diabetes in a Latino population. JAMA. 2014;311:2305–14.
    1. Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014;343:80–4.
    1. Majithia AR, et al. Rare variants in PPARG with decreased activity in adipocyte differentiation are associated with increased risk of type 2 diabetes. Proc Natl Acad Sci U S A. 2014;111:13127–32.
EXTENDED METHODS REFERENCES
    1. Guey LT, et al. Power in the phenotypic extremes: a simulation study of power in discovery and replication of rare variants. Genet Epidemiol. 2011;35:236–46.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–60.
    1. DePristo MA, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43:491–8.
    1. McKenna A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–303.
    1. Jun G, et al. Detecting and estimating contamination of human DNA samples in sequencing and array-based genotype data. Am J Hum Genet. 2012;91:839–48.
    1. Abecasis GR, et al. An integrated map of genetic variation from 1,092 human genomes. Nature. 2012;491:56–65.
    1. Handsaker RE, Korn JM, Nemesh J, McCarroll SA. Discovery and genotyping of genome structural polymorphism by sequencing on a population scale. Nat Genet. 2011;43:269–76.
    1. Browning SR, Browning BL. Rapid and accurate haplotype phasing and missing-data inference for whole-genome association studies by use of localized haplotype clustering. Am J Hum Genet. 2007;81:1084–97.
    1. Li Y, Sidore C, Kang HM, Boehnke M, Abecasis GR. Low-coverage sequencing: implications for design of complex trait association studies. Genome Res. 2011;21:940–51.
    1. Price AL, et al. Long-range LD can confound genome scans in admixed populations. Am J Hum Genet. 2008;83:132–5. author reply 135-9.
    1. Weale ME. Quality control for genome-wide association studies. Methods Mol Biol. 2010;628:341–72.
    1. Wellcome Trust Case Control Consortium Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–78.
    1. Price AL, et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38:904–9.
    1. Fuchsberger C, Abecasis GR, Hinds DA. minimac2: faster genotype imputation. Bioinformatics. 2015;31:782–4.
    1. Firth D. Bias reduction of maximum-likelihood-estimates. Biometrika. 1993;80:27–38.
    1. Ma C, Blackwell T, Boehnke M, Scott LJ, GoT2D investigators Recommended joint and meta-analysis strategies for case-control association testing of single low-count variants. Genet Epidemiol. 2013;37:539–50.
    1. Morris AP. Transethnic meta-analysis of genomewide association studies. Genet Epidemiol. 2011;35:809–22.
    1. Seldin MF, Pasaniuc B, Price AL. New approaches to disease mapping in admixed populations. Nat Rev Genet. 2011;12:523–8.
    1. Price AL, et al. Sensitive detection of chromosomal segments of distinct ancestry in admixed populations. PLoS Genet. 2009;5:e1000519.
    1. Churchhouse C, Marchini J. Multiway admixture deconvolution using phased or unphased ancestral panels. Genet Epidemiol. 2013;37:1–12.
    1. Purcell SM, et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature. 2014;506:185–90.
    1. Lee S, Wu MC, Lin X. Optimal tests for rare variant effects in sequencing association studies. Biostatistics. 2012;13:762–75.
    1. Marchini J, Howie B, Myers S, McVean G, Donnelly P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat Genet. 2007;39:906–13.
    1. Devlin B, Roeder K. Genomic control for association studies. Biometrics. 1999;55:997–1004.
    1. Willer CJ, Li Y, Abecasis GR. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics. 2010;26:2190–1.
    1. Hindorff LA, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A. 2009;106:9362–7.
    1. Korn JM, et al. Integrated genotype calling and association analysis of SNPs, common copy number polymorphisms and rare CNVs. Nat Genet. 2008;40:1253–60.
    1. Rice WR. A Consensus Combined P-Value Test and the Family-Wide Significance of Component Tests. Biometrics. 1990;46:303–308.
    1. Yang J, et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat Genet. 2012;44:369–75. S1–3.
    1. Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet. 2011;88:76–82.
    1. Harrow J, et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 2012;22:1760–74.
    1. Ernst J, Kellis M. Discovery and characterization of chromatin states for systematic annotation of the human genome. Nat Biotechnol. 2010;28:817–25.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50.
    1. Lage K, et al. A human phenome-interactome network of protein complexes implicated in genetic disorders. Nat Biotechnol. 2007;25:309–16.
    1. Nepusz T, Yu H, Paccanaro A. Detecting overlapping protein complexes in protein-protein interaction networks. Nat Methods. 2012;9:471–2.
    1. Jia P, Zheng S, Long J, Zheng W, Zhao Z. dmGWAS: dense module searching for genome-wide association studies in protein-protein interaction networks. Bioinformatics. 2011;27:95–102.
    1. Lambert BW, Terwilliger JD, Weiss KM. ForSim: a tool for exploring the genetic architecture of complex traits with controlled truth. Bioinformatics. 2008;24:1821–2.
    1. Eyre-Walker A. Evolution in health and medicine Sackler colloquium: Genetic architecture of a complex trait and its implications for fitness and genome-wide association studies. Proc Natl Acad Sci U S A. 2010;107(Suppl 1):1752–6.
    1. Lyssenko V, et al. Clinical risk factors, DNA variants, and the development of type 2 diabetes. N Engl J Med. 2008;359:2220–32.

Source: PubMed

3
구독하다