Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 7.
Published in final edited form as: Nat Genet. 2012 Oct 28;44(12):1294–1301. doi: 10.1038/ng.2435

Bayesian refinement of association signals for 14 loci in 3 common diseases

The Wellcome Trust Case Control Consortium, Julian B Maller 1,2, Gilean McVean 1,2, Jake Byrnes 1, Damjan Vukcevic 1, Kimmo Palin 3, Zhan Su 1, Joanna M M Howson 4,5, Adam Auton 1, Simon Myers 1,2, Andrew Morris 1, Matti Pirinen 1, Matthew A Brown 6,7, Paul R Burton 8,9, Mark J Caulfield 10, Alastair Compston 11, Martin Farrall 12, Alistair S Hall 13, Andrew T Hattersley 14,15, Adrian V S Hill 1, Christopher G Mathew 16, Marcus Pembrey 17, Jack Satsangi 18, Michael R Stratton 3,19, Jane Worthington 20, Nick Craddock 21, Matthew Hurles 3, Willem Ouwehand 3,22,23, Miles Parkes 24, Nazneen Rahman 19, Audrey Duncanson 25, John A Todd 5, Dominic P Kwiatkowski 1,3, Nilesh J Samani 26,27, Stephen C L Gough 28,29, Mark I McCarthy 1,28,29, Panagiotis Deloukas 3, Peter Donnelly 1,2
PMCID: PMC3791416  EMSID: EMS51446  PMID: 23104008

Abstract

To further investigate susceptibility loci identified by genome-wide association studies, we genotyped 5,500 SNPs across 14 associated regions in 8,000 samples from a control group and 3 diseases: type 2 diabetes (T2D), coronary artery disease (CAD) and Graves’ disease. We defined, using Bayes theorem, credible sets of SNPs that were 95% likely, based on posterior probability, to contain the causal disease-associated SNPs. In 3 of the 14 regions, TCF7L2 (T2D), CTLA4 (Graves’ disease) and CDKN2A-CDKN2B (T2D), much of the posterior probability rested on a single SNP, and, in 4 other regions (CDKN2A-CDKN2B (CAD) and CDKAL1, FTO and HHEX (T2D)), the 95% sets were small, thereby excluding most SNPs as potentially causal. Very few SNPs in our credible sets had annotated functions, illustrating the limitations in understanding the mechanisms underlying susceptibility to common diseases. Our results also show the value of more detailed mapping to target sequences for functional studies.


Genome-wide association studies (GWAS) have had unprecedented success in identifying genomic regions and candidates for causal genes where genetic variation, namely SNP markers, is most strongly associated with phenotypic variation and disease susceptibility. To date, over 1,500 such regions have been identified across more than 200 diseases and phenotypes (Catalog of Published Genome-Wide Association Studies; see URLs). By comparing results and candidate genes across multiple GWAS regions, these studies can indicate new candidates for causal pathways, such as autophagy in Crohn’s disease1, triggering a new wave of functional studies and providing genetic validation of therapeutic strategies1. Even though GWAS genotyping chips, including the Affymetrix Human 500K chip we used in an initial GWAS of seven common diseases2, provide very good mapping coverage of common variation of the genome in Europeans, it is possible that with a much denser set of SNP markers we can refine association signals to particular candidate genes or even detect missing disease association signals that were poorly tagged on the GWAS chip.

It is now appreciated that most common variants that are associated with altered disease risk do not have obvious functional consequences, leading to the plausible conclusion that they affect the regulation of gene expression in some way. Nevertheless, most cis-acting regulatory variation lies within or very proximal to the structural genes3 affected by those functional polymorphisms, and, hence, identifying all or most of the SNPs with the strongest disease associations within a region will help to identify candidate genes with greater confidence. This follow-up genotyping, often referred to as fine mapping, can identify new candidate genes and causal variants in GWAS-identified regions, for example, the nonsynonymous SNP rs3184504 in SH2B3 in type 1 diabetes4. In addition, it can lead to the identification of regions and SNPs that are highly unlikely to be causal. In the present study, as a follow-up to the Wellcome Trust Case Control Consortium (WTCCC) GWAS2, we studied 14 associated regions in 3 diseases, T2D, CAD and Graves’ disease, by attempting to genotype all known SNPs in the associated regions. In five of these regions, we had undertaken SNP discovery through resequencing of controls. As a result, we not only have refined some of the association signals to certain genes and SNPs, but we have also developed and applied an informative way of analyzing and interpreting the results of dense SNP mapping using a Bayesian approach.

RESULTS

Experimental design

We genotyped all known SNPs for which we could design assays across 13 GWAS-associated regions using an Illumina iSELECT assay. These were typed on 7,894 samples in total: 1,930 common controls and ~2,000 cases each with T2D, CAD and the common autoimmune thyroid disease, Graves’ disease. The three diseases were chosen to reflect a range of putative disease etiologies, and the regions were selected to include both strong and weaker signals in our original GWAS. A breakdown of the numbers of SNPs and their sources is given in Table 1. Our fine-mapping experiment was preceded by resequencing of 32 unrelated Utah residents of Northern and Western European ancestry (CEU) HapMap individuals across 5 of the fine-mapping regions (and 11 other regions showing association in WTCCC-studied diseases), to identify putative causal mutations in these regions; this information added to existing SNP variation resources. Further details of these resequencing experiments are given in Supplementary Figures 17, Supplementary Tables 1 and 2 and the Supplementary Note.

Table 1. Region definitions with SNP counts and sources.

Affymetrix 500K
HapMap
Resequencing
DbSNP
Phenotype Chr. Start
(Mb)
End
(Mb)
Gene Total Genotyped Passed
quality
control
Polymorphic Total Genotyped Passed
quality
control
Polymorphic Total Genotyped Passed
quality
control
Polymorphic Total Genotyped Pass QC Polymorphic
CAD 1 109.54 109.65 SORT1 49 19 17 13 76 75 50 50 141 77 42 22 539 239 173 127
9 21.92 22.13 CDKN2A-CDKN2B 107 40 37 30 176 174 128 128 691 443 184 64 984 464 314 206
10 44.01 44.15 CXCL12 115 42 40 33 161 159 111 111 74 35 23 16 957 416 303 238
1 220.78 221.04 lq41 68 23 23 22 11 11 6 6 109 56 28 25 970 399 305 266
2 226.73 226.91 2q36 67 25 24 18 100 96 72 72 378 216 121 41 691 330 223 138
T2D 16 52.35 52.41 FTO 42 15 14 13 75 74 53 52 356 199 98 59 313 128 97 88
10 94.19 94.49 HHEX 48 18 16 14 152 145 115 114 974 564 248 162 1,019 457 309 253
6 20.63 20.84 CDKAL1 84 30 28 26 241 229 167 167 573 364 135 74 1,115 503 347 265
10 114.71 114.82 TCF7L2 42 14 14 14 53 53 34 34 75 32 24 19 369 159 118 92
7 28.01 28.23 JAZF1 101 37 34 30 157 152 104 104 63 34 15 14 873 369 290 214
Graves’
disease
2 204.38 204.53 CTLA-4 91 35 33 23 135 133 104 104 59 33 13 13 754 322 246 186
10 6.06 6.17 CD25-IL2RA 97 38 32 27 111 111 71 71 17 13 2 2 1,222 566 391 265
1 155.81 156.09 FCRL3 125 46 44 35 206 200 131 130 43 28 10 5 1,637 743 552 342
Total 1,036 382 356 298 1,654 1,612 1,146 1,143 3,553 2,094 943 516 11,443 5,095 3,668 2,680

Chr., chromosome. Genomic coordinates are from NCBI Build 36, and references to dbSNP correspond to Build 128. The numbers of SNPs for each region come from various sources: the Affymetrix 500K genotyping chip; HapMap 2; targeted resequencing experiments (either within WTCCC, provided by collaborators or through mining of the Watson and Venter genomes); and dbSNP. Each SNP was attributed to the left-most source in which it was found. For each SNP source, counts are shown for the total number of SNPs, the number for which successful genotyping assays were designed and performed, the number that passed quality control filters and the number that were polymorphic in our samples (MAF > 0.01).

Overall findings

In the subsequent analyses for a particular disease, we leveraged our design by comparing cases for that disease against an expanded reference group consisting of the common controls and the cases with unrelated diseases: Graves’ disease cases were compared against common controls and T2D and CAD cases, and T2D and CAD cases were each compared against common controls and Graves’ disease cases. Comparing cases only against the common controls resulted in weaker signals, as expected, but no change in the pattern of signals (data not shown). One region on chromosome 9p21, containing CDKN2A-CDKN2B, is associated with both CAD and T2D, such that there were 14 association regions in total analyzed in the fine-mapping experiment.

We developed and applied a new Bayesian method for the statistical analysis of the fine-mapping data, which seems to have several advantages over other approaches. In the Bayesian framework, the evidence for association at a SNP is measured by the Bayes factor. Under certain assumptions, these Bayes factors can be used to calculate the posterior probability for each SNP, that is, the probability when taking into consideration the fine-mapping data that the SNP is driving the association signal (Online Methods). These posterior probabilities can be directly compared between SNPs within and across regions, in a way that is not straightforward, for example, with association P values.

For each region, the posterior probability associated with each SNP can be calculated (Fig. 1 and Supplementary Fig. 8). Uncertainty after fine mapping as to the identity of the causal SNPs is conveniently handled by describing credible sets. In each region, one can choose the smallest set of SNPs accounting for 95% or 99% of the posterior probability. These credible sets are somewhat analogous to confidence intervals: in a particular region, if the true causal SNP has been genotyped, we can be reasonably confident that it will be in the relevant credible set. If the credible set for a particular region contains a small number of SNPs, the fine-mapping experiment has been informative in narrowing down the set of SNPs that might be causal. In the other direction, SNPs not contained in the credible set can be excluded from being responsible for the primary association signal, such that, if the credible set is large, the fine-mapping experiment has added little or no resolution in the search for the causal variant(s).

Figure 1.

Figure 1

Association plots showing the signal strength in each region as the posterior probability of each SNP passing quality control. The estimated recombination rate is shown in blue (right y axis). Genomic position is shown on the x axis with Human Genome Build 36 coordinates. SNPs are colored according to membership in credible sets: yellow, 95% credible set; purple, 99% credible set; gray outline, neither set. Genes in the region are shown at the bottom in green. For each plot, the region name, phenotype and sizes of the 95% and 99% credible sets are indicated. All SNPs in the 95% credible set are a subset of the 99% credible set. SNPs with large posterior probabilities represent those most likely to be causal among the SNPs typed.

Our results identify several features. First, there are only three regions in which much of the posterior probability after the fine mapping rests on a single SNP: TCF7L2 in T2D, CTLA4 in Graves’ disease and CDKN2A-CDKN2B in T2D (Fig. 1). The finding at the third region is misleading, as there is a substantially stronger signal generated by pairs of SNPs at CDKN2A-CDKN2B in T2D (Supplementary Note). Second, in 7 of the 14 regions (CDKN2A-CDKN2B in CAD, CTLA4 in Graves’ disease and CDKN2A-CDKN2B, CDKAL1, FTO, HHEX and TCF7L2 in T2D), including the 3 just mentioned, the credible sets are relatively small, including fewer than 34 SNPs, and the fine-mapping experiment has provided useful information, at least in excluding large numbers of SNPs from being causal (Supplementary Table 3). In the other 7 regions, 1q41, 2q36, IL2RA, CXCL12, FCRL3, JAZF1 and SORT1, the project has not excluded many of the SNPs as potentially being causal (or being good markers)—for each region, the 95% credible set contains at least 75 and typically over 100 SNPs. Nonetheless, two of these seven regions (SORT1 and IL2RA) have the property that, even though large numbers of SNPs cannot be excluded, substantial posterior weight rests on one or a few SNPs, and, therefore, typing additional samples in these regions should lead to a much smaller credible set.

The seven regions for which our additional genotyping did not eliminate many of the SNPs are distinguished by the fact that they were the regions where the initial association signal in our data was weakest. Our results indicate that, in these regions, these smaller signals are due to the causal variant(s) having only very modest effect sizes, explaining the difficulty in fine mapping and, therefore, requiring much larger sample sizes to begin to resolve the linkage disequilibrium in the regions and refine the associations. There was the possibility of a considerably stronger signal at one of the new SNPs genotyped if one of the new SNPs was a causal variant of large effect that was tagged only poorly in the original GWAS or a good tag marker for such a variant. We note that none of the 14 regions showed evidence of such a variant.

Imputation

Although we aimed to genotype all known SNPs in the regions at the time that the experiment was designed, one limitation of our study was that the catalog of variation was not complete. In addition, we were only able to design assays for 78% of SNPs, and only obtained genotypes that successfully passed quality control for 94% of the SNPs for which assays were designed. To recover information for SNPs not genotyped in our experiment, we also imputed genotypes at all SNPs present in 1000 Genomes Project data (June 2011 release) for which we did not have genotype data (Supplementary Note). The resulting association plots are shown in Supplementary Figure 9. The signal at the top genotyped SNP is compared with that at the top imputed SNP in Supplementary Table 4. There was only one region, 2q36 in CAD, where the top SNP after imputation was not one of the genotyped SNPs, and, even in this region, imputation did not greatly affect the overall results: the top imputed SNP had a posterior weight of 0.051, only slightly greater than the weight (0.049 in the imputed data set) for the top genotyped SNP, and the size of the 95% (99%) credible region increased from 86 (99) SNPs to 107 (122). No imputed SNP had higher posterior probability than the top genotyped SNP in any other region, and the imputed SNPs did not change the overall results in these regions, either by identifying any 1000 Genomes Project SNPs with large signals in the seven regions where our additional genotyping did not exclude many of the SNPs or by greatly changing the composition of the credible sets in the regions where our efforts were informative.

Mapping information gained

We can also ask whether the fine-mapping effort added to our understanding from the original GWAS study. The evidence for association of the top SNP after the fine-mapping experiment was compared with that from the top SNP in the region on each of three commercial geno-typing chips (Affymetrix Human 500K SNP array and Illumina 550K and 1.2M SNP arrays) (Table 2). The Bayes factor ratio allows a direct comparison of the relative evidence for association of the top SNP from a particular chip to the top SNP from our new results: for example, a Bayes factor ratio of 0.22 means that the posterior probability for the top SNP from the chip is smaller by a factor of 0.22 than that for the new top SNP. The new top SNP improved on the top SNP from the Affymetrix 500K and Illumina 550K and 1.2M arrays, respectively, in 13, 12 and 8 of the 14 regions, with comparisons among SNP chips differing across regions. Our results, not unexpectedly, indicate that the top GWAS SNPs are typically not the best markers for the causal variant. Another comparison of GWAS results and our findings is given in Supplementary Table 5. For the Affymetrix 500K array, we show how many of the SNPs in the credible set after the fine-mapping experiment were typed in the original GWAS. In most regions, the vast majority of the SNPs in credible sets after the fine-mapping experiment were not typed in the original association study.

Table 2. Comparison of association evidence between top SNPs after fine mapping and top SNPs on GWAS chips.

Disease Region SNP set SNP ID Risk allele
frequency
(controls)
Risk
allele
Effect size
(relative risk)
log Bayes
factor
Bayes
factor ratio
λ s
CAD SORT1 Genotyped in fine
mapping
rs3832016 0.79 A 1.22 (1.11–1.35) 2.68 1.00 1.006
Affymetrix 500K rs599839 0.78 A 1.17 (1.07–1.29) 1.59 0.08 1.004
Illumina 550K rs611917 0.69 A 1.17 (1.07–1.27) 1.96 0.19 1.005
Illumina 1.2M rs660240 0.79 G 1.22 (1.10–1.34) 2.54 0.72 1.006
CDKN2A-CDKN2B Genotyped in fine
mapping
rs1537370 0.47 A 1.40 (1.29–1.51) 14.03 1.00 1.028
Affymetrix 500K rs6475606 0.47 A 1.40 (1.29–1.51) 13.95 0.83 1.028
Illumina 550K rs10116277 0.47 A 1.40 (1.29–1.51) 13.99 0.92 1.028
Illumina 1.2M rs1537370 0.47 A 1.40 (1.29–1.51) 14.03 1.00 1.028
CXCL12 Genotyped in fine
mapping
rs34161818 0.84 A 1.21 (1.08–1.35) 1.56 1.00 1.004
Affymetrix 500K rs977754 0.85 A 1.18 (1.06–1.33) 1.16 0.39 1.003
Illumina 550K rs977754 0.85 A 1.18 (1.06–1.33) 1.16 0.39 1.003
Illumina 1.2M rs977754 0.85 A 1.18 (1.06–1.33) 1.16 0.39 1.003
1q41 Genotyped in fine
mapping
rs2936023 0.85 T 1.21 (1.08–1.36) 1.58 1.00 1.004
Affymetrix 500K rs17464857 0.85 A 1.17 (1.05–1.31) 0.93 0.22 1.003
Illumina 550K rs17464857 0.85 A 1.17 (1.05–1.31) 0.93 0.22 1.003
Illumina 1.2M rs11485123 0.85 G 1.17 (1.05–1.31) 0.93 0.22 1.003
2q36 Genotyped in fine
mapping
rs2673145 0.41 A 1.20 (1.11–1.30) 3.76 1.00 1.008
Affymetrix 500K rs2943646 0.64 G 1.19 (1.10–1.29) 3.01 0.18 1.007
Illumina 550K rs2972153 0.67 G 1.21 (1.11–1.32) 3.45 0.49 1.008
Illumina 1.2M rs2972153 0.67 G 1.21 (1.11–1.32) 3.45 0.49 1.008
T2D FTO a Genotyped in fine
mapping
rs17817449 0.40 C 1.26 (1.17–1.36) 6.69 1.00 1.013
Combined rs17817449,rs8063946 1.018
Affymetrix 500K rs8050136 0.40 A 1.26 (1.17–1.36) 6.49 0.62 1.013
Illumina 550K rs8050136 0.40 A 1.26 (1.17–1.36) 6.49 0.62 1.013
Illumina 1.2M rs17817449 0.40 C 1.26 (1.17–1.36) 6.69 1.00 1.013
CDKN2A-CDKN2B a Genotyped in fine
mapping
rs12555274 0.23 C 1.26 (1.15–1.37) 4.75 1.00 1.011
Combined rs10811661,rs10217762 1.012
Affymetrix 500K rs10811661 0.83 A 1.27 (1.14–1.41) 3.46 0.05 1.007
Illumina 550K rs2383208 0.82 A 1.26 (1.13–1.40) 3.20 0.03 1.007
Illumina 1.2M rs2383208 0.82 A 1.26 (1.13–1.40) 3.20 0.03 1.007
HHEX Genotyped in fine
mapping
rs10882098 0.59 G 1.21 (1.12–1.31) 4.01 1.00 1.009
Affymetrix 500K rs5015480 0.59 G 1.20 (1.11–1.30) 3.80 0.61 1.008
Illumina 550K rs5015480 0.59 G 1.20 (1.11–1.30) 3.80 0.61 1.008
Illumina 1.2M rs5015480 0.59 G 1.20 (1.11–1.30) 3.80 0.61 1.008
CDKAL1 a Genotyped in fine
mapping
rs7756992 0.27 G 1.29 (1.19–1.40) 6.71 1.00 1.014
Combined rs7756992,rs6456360 1.021
Affymetrix 500K rs9460546 0.31 C 1.25 (1.15–1.35) 5.38 0.05 1.012
Illumina 550K rs7756992 0.27 G 1.29 (1.19–1.40) 6.71 1.00 1.014
Illumina 1.2M rs7756992 0.27 G 1.29 (1.19–1.40) 6.71 1.00 1.014
TCF7L2 Genotyped in fine
mapping
rs7903146 0.30 A 1.40 (1.29–1.52) 13.61 1.00 1.027
Affymetrix 500K rs4506565 0.32 T 1.37 (1.27–1.49) 12.24 0.04 1.024
Illumina 550K rs7903146 0.30 A 1.40 (1.29–1.52) 13.61 1.00 1.027
Illumina 1.2M rs7903146 0.30 A 1.40 (1.29–1.52) 13.61 1.00 1.027
JAZF1 Genotyped in fine
mapping
rs12531540 0.51 G 1.14 (1.06–1.23) 1.75 1.00 1.004
Affymetrix 500K rs1859687 0.06 C 1.25 (1.08–1.45) 1.12 0.23 1.003
Illumina 550K rs498475 0.37 G 1.14 (1.05–1.23) 1.44 0.49 1.004
Illumina 1.2M rs498475 0.37 G 1.14 (1.05–1.23) 1.44 0.49 1.004
Graves’ disease CTLA4 Genotyped in fine
mapping
rs11571297 0.51 A 1.39 (1.29–1.50) 16.08 1.00 1.027
Affymetrix 500K rs3087243 0.55 G 1.38 (1.28–1.49) 14.89 0.06 1.025
Illumina 550K rs231804 0.57 A 1.37 (1.27–1.48) 13.58 0.00 1.023
Illumina 1.2M rs11571291 0.58 A 1.38 (1.28–1.48) 13.87 0.01 1.024
CD25-IL2RA Genotyped in fine
mapping
rs10905669 0.23 A 1.20 (1.10–1.30) 3.10 1.00 1.007
Affymetrix 500K rs10905669 0.23 A 1.20 (1.10–1.30) 3.10 1.00 1.007
Illumina 550K rs7090530 0.60 A 1.16 (1.08–1.25) 2.49 0.25 1.005
Illumina 1.2M rs10905669 0.23 A 1.20 (1.10–1.30) 3.10 1.00 1.007
FCRL3 Genotyped in fine
mapping
rs11264798 0.52 C 1.17 (1.09–1.26) 3.00 1.00 1.006
Affymetrix 500K rs2785663 0.58 C 1.17 (1.08–1.26) 2.80 0.63 1.006
Illumina 550K rs2785665 0.58 A 1.17 (1.08–1.26) 2.76 0.58 1.006
Illumina 1.2M rs11264798 0.52 C 1.17 (1.09–1.26) 3.00 1.00 1.006

For each region, shown is the top SNP (based on Bayes factor) after the fine-mapping experiment among various sets of SNPs, including SNPs passing quality control in the fine-mapping experiment and SNPs from the Affymetrix Human 500K SNP array, the Illumina 550K SNP array and the Illumina 1.2M SNP array. Shown are risk allele frequency, risk allele, relative risk (and 95% confidence interval (CI)), log10 of the Bayes factor for the SNP, the ratio of the Bayes factor for the SNP to the Bayes factor for the top fine-mapping SNP and the contribution of that SNP to the sibling relative risk (λs).

a

For the three regions with evidence for a second associated SNP, the combined contribution of the pair of SNPs to the sibling relative risk is shown.

The new top SNPs do not greatly change the proportion of heritability explained by the locus (Table 2). There can be strong evidence in favor of one SNP over another, even when there are only slight differences in effect sizes (most notably, in CTLA4 in Graves’ disease, but similar results were also found in several other SNP comparisons) and, hence, only slight differences in heritability explained by the locus.

Bioinformatic annotation

In most of the regions where the fine mapping considerably refined the association signal, there was still a set of correlated SNPs that could not be separated on the basis of our current genetic data in terms of the possibility that a SNP might be functional and causal. We therefore cross-referenced our most credible SNPs against all relevant annotation tracks found in the UCSC Genome Browser. We first considered annotations based on primary DNA sequence, before turning to more recent tissue-specific annotations relating principally to properties of chromatin.

The results for annotations based primarily on DNA sequence for the seven regions where the fine mapping excluded most SNPs in the region are shown in Table 3. The lack of putative functional annotations is noteworthy. Of the 109 (247) SNPs in the 95% (99%) credible sets across the 7 regions, there were no SNPs in coding exons and no (5) SNPs in 5′ and 3′ RefGene UTRs. Two SNPs were found in the CDKN2B-AS1 gene (also known as ANRIL) for a non-coding antisense RNA, but they each had very low posterior probability. Many SNPs were intronic, but none of these were in canonical splice sites. Cross-referencing our SNPs against other gene annotation tracks, including non-coding and microRNA (miRNA) tracks, did not indicate that any other SNPs were genic.

Table 3. Biological annotations for the 109 and 247 SNPs making up the 95% and 99% credible sets, respectively, across 7 fine-mapped regions.

Proportion of SNPs in
credible set
Proportion of posterior
probability in credible set

Annotation typea Annotation 95% 99% 95% 99%
dbSNP130 functions Nonsynonymous 0 0 0 0
Synonymous 0 0 0 0
Intron 0.67 0.53 0.43 0.43
Splicing 0 0 0 0
5′ UTR 0 0 0 0
3′ UTR 0 0.02 0 0
Unknown (intergenic) 0.31 0.44 0.56 0.56
Other gene prediction methods Alternative splicing events 0 0 0 0
Non-coding RNA 0 0 0 0
miRNA (miRBase) 0 0 0 0
ChIP-seq transcription factor–binding site summary ENCODE ChIP-seq transcription factor–binding sites 0.23 0.32 0.21 0.20
DNase I hypersensitivity Duke/UW DNase I hypersensitivity (P < 0.05) 0.10 0.11 0.07 0.08
UW nucleosome occupancy (A375 >1.0) 0.01 0.03 0 0
UW nucleosome occupancy (dennis >1.0) 0.03 0.04 0.02 0.02
UW nucleosome occupancy (mec < −1.0) 0.01 0.01 0 0
Other regulatory predictions tfbsCons 0.01 0.01 0 0
miRNA target site 0 0 0 0
7× reg score >0.1 0.14 0.1 0.07 0.07
Sequence conservation 17-way most conserved vertebrate 0.05 0.04 0.02 0.02
28-way most conserved mammal 0.05 0.03 0.02 0.02

UW, University of Washington.

a

See the supplementary Note for details on specific annotation classes.

Next, we examined recently derived annotations associated with histone modification and chromatin accessibility2. This analysis was complicated by several factors. The first is that these domains tend to be modified in a tissue-specific manner. Relevant tissues for particular diseases are not always clear and are often not unique, and data for these tissues may in any event not be available. In addition, the precision of these annotations is sometimes either limited or unclear. Overall summaries of the annotations are provided in the Supplementary Table 6 and the Supplementary Note, but, at what seems to be an early stage in understanding the mechanisms involved in chromatin accessibility, this analysis did not provide compelling information to distinguish SNPs highlighted in the fine-mapping experiment.

Secondary signals

In addition to single-SNP analyses, in each region, we performed conditional and other analyses to look for secondary signals. There are several regions with clear evidence for secondary signals (CDKN2A-CDKN2B, FTO and CDKAL1; Supplementary Note). We also assessed the best model for relating SNP genotype to disease risk. For most reported GWAS associations, there is no significant evidence for departure from the simple model in which each additional copy of the risk allele increases disease risk by the same multiplicative factor. In this simple model, the log odds of disease increase in an additive manner with each additional copy of the risk allele, such that the model is sometimes referred to as the additive model. But, as has been noted elsewhere5, the power to detect departures from this model is limited unless the true causal variant or a SNP highly correlated with it is typed in the study. With the much more extensive genotyping in this study, it is natural to revisit this question. We found no compelling evidence for departures from the additive model.

Detailed results for the 14 regions studied are described in the Supplementary Note (see also Supplementary Figs. 821 and Supplementary Tables 313).

DISCUSSION

We have found that the use of Bayesian statistical approaches for assigning posterior probabilities and credible sets of SNPs is informative in refining the association signals from GWAS-detected loci with denser genotyping. The calculated value of the posterior probability depends on a number of assumptions, including the assumption that there is a single causal SNP that is typed in the study. This will not always be the case, and, indeed, our data show this is not true for several of the regions we studied, most notably CDKN2A-CDKN2B in T2D (Supplementary Note). Nonetheless, the ratio of these posterior probabilities for a pair of SNPs reduces to the ratio of the Bayes factors for the pair, and, in a Bayesian framework, this ratio is the quantity that summarizes the weight of evidence in the data for one SNP compared to the other (as shown, for example, in Table 2). The calculated posterior probabilities can be thought of as the ratio of the evidence for the particular SNP compared to that for all SNPs typed, such that they are helpful summaries of the overall evidence for each individual SNP based on the fine-mapping data, whether or not there is a single causal SNP or a very good marker typed in the study.

In addition to the detailed analyses of particular regions (Supplementary Note), there are several general conclusions from our fine-mapping experiments. First, it seems that, in general, weaker GWAS signals are not driven by poor tagging of a variant with a larger effect size but simply by genuinely smaller effects. Our fine-mapping experiment was powered to find larger effect variants that were genotyped or tagged well, but such variants were not identified in any region with a relatively small effect in the original GWAS. Consequently and because the required sample size depends on the true effect size, in these regions, our additional genotyping did not add greatly to the resolution of the signal for experiments of our size (2,000 cases and, in effect, 4,000 or 6,000 controls). For smaller effect sizes, much larger sample sets are required, and, even then, if linkage disequilibrium is very strong and extended, very large sample sizes can still be uninformative.

Evidence for the involvement of multiple SNPs in a single region was obtained in 3 of the 14 regions, even though our sample size for each disease was small. It thus seems likely that allelic heterogeneity is not uncommon at GWAS loci, and this knowledge will not only contribute to the evidence for a particular candidate gene but will also provide an allelic series to aid functional studies and help explain the familial clustering of the disease. Taken together, the pairs of SNPs at these three loci substantially increase the strength of the genetic effect and also the heritability explained by the locus (Table 2).

One limitation with our study is that we were restricted to genotyping only those SNPs known at the time of the study design and our specific sequencing efforts, and we therefore could simply have missed the causal variants or the most associated SNP markers, particularly at lower minor allele frequencies (MAFs). However, we used data from the 1000 Genomes Project to assess our coverage of variants in the regions studied (Table 4). For example, in the June 2011 1000 Genomes Project data release, there were 1,920 and 1,601 SNPs in samples from individuals of European descent in our fine-mapping regions with MAFs from 1–2% and 2–4%, respectively. Of these, we directly typed 92 (4.8%) and 240 (15%), respectively, and, in addition, obtained good-quality imputation data on a further 746 and 1,065 SNPs. Our project typed a much denser set of SNPs than GWAS chips, leading to improved imputation. Thus, in these MAF ranges, we obtained actual or well-imputed data on 44% and 82% of these variants. For all higher MAF ranges, the proportion captured or well imputed was at least 85% (Table 4). Coverage via genotyping or imputation of variants with MAF of <1% was much lower, but we do not believe this undermines our main conclusions (Supplementary Note).

Table 4. Proportion of 1000 Genomes Project sNPs captured in this experiment (by direct genotyping or imputation) stratified by MAF.

MAF Genotyped in
fine mapping
Genotyped
or well imputeda
1000 Genomes
Project
Proportion
captured
MAF < 0.01 125 1,665 20,306 (12,263)b 0.082 (0.14)c
0.01 ≤ MAF < 0.02 92 838 1,920 0.44
0.02 ≤ MAF < 0.04 240 1,305 1,601 0.82
0.04 ≤ MAF < 0.06 256 675 794 0.85
0.06 ≤ MAF < 0.10 260 808 901 0.90
0.10 ≤ MAF < 0.20 614 972 1,083 0.90
0.20 ≤ MAF 1,784 2,824 3,196 0.88

Imputation and MAFs are based on a reference panel of 143 European haplotypes in the 1000 Genomes Project June 2011 data release. See the Supplementary Note for imputation details.

a

SNPs were deemed to be well imputed if average maximum posterior (as returned by IMPUTE1) was greater than 0.98, the IMPUTE1 info score was above 0.8 and there was less than 2% missing data.

b

Count of SNPs with at least two observations of the minor allele in the reference panel.

c

Proportion of SNPs with at least two observations of the minor allele in the reference panel.

Ranking of SNPs by posterior probability is in general different from ranking based on P values, complicating direct comparisons of our findings with those from other studies. In comparing our top SNPs with those in a recent large follow-up study of T2D6, we found that the top SNPs were identical in three regions (CDKN2A-CDKN2B, CDKAL1 and TCF7L2) and almost perfect proxies in the other two regions for which our experiment was informative (FTO, r2 = 0.995; HHEX, r2 = 0.967) (Supplementary Table 9; this table also compares our findings with those described in a recent review of the GWAS results for CAD7, for which comparison is complicated because our top SNPs were not necessarily typed in the studies reviewed).

This study provides a statistical platform for intuitive and informative analysis and interpretation of GWAS and denser mapping data. We have succeeded via resequencing, dense genotyping and statistical analysis in substantially refining the association signals at several risk-associated loci. This fundamental knowledge will aid future functional studies of specific fine-mapped sequences and candidate genes, help explain heritability and lead to the identification of functional, causal pathways in disease that could be safely modulated for the prevention or intervention of common and rare diseases.

ONLINE METHODS

Bayesian approach

We adopted a Bayesian statistical perspective for analysis, with the strength of evidence for association measured via the Bayes factor for each SNP: the probability of the genotype configuration at that SNP in cases and controls under the alternative hypothesis that the SNP is associated with disease status divided by the probability of the genotype configuration at that SNP in cases and controls under the null hypothesis that disease status is independent of genotype at that SNP (Supplementary Note). Large values of the Bayes factor correspond to strong evidence for association, in the way that small P values would correspond to strong evidence in a frequentist perspective.

One advantage of the Bayesian perspective is that Bayes factors for different SNPs can be compared quantitatively, which does not seem straightforward with P values. For example, for a particular region, under specific assumptions about how many causal SNPs are in the set of genotyped SNPs, it is straightforward to calculate the posterior probability that any particular SNP is causal taking into consideration data from the fine-mapping experiment. For definiteness, we calculate these posterior probabilities under the simple assumption that exactly one of the genotyped SNPs in each region is causal and that it is equally likely, a priori, to be any of the genotyped SNPs in the region. We cannot know that these previous assumptions are true for any particular region, and they may well not be, but the resulting probabilities can be thought of as the relative strength of evidence in favor of each of the SNPs studied. Defining BFk as the Bayes factor for SNP k, we show in the Supplementary Note that the posterior probably for SNP k is equal to

BFkjBFj

where j indexes SNPs in the region. Regardless of whether the causal SNP or SNPs have been typed in the fine-mapping experiment, SNPs with a low value for this posterior probability are unlikely to be causal.

SNP selection, genotyping and quality control

The case samples used in this study were as previously described in earlier WTCCC studies2,8. Control samples were a subset of those used in the original WTCCC study2.

The boundaries of the genomic regions for study were determined as follows. We took the top associated SNP in the region from published results at the time of study design (referred to here as the ‘focal SNP’) and extended the region upstream and downstream by a genetic distance of 0.1 cM, using HapMap fine-scale estimates of recombination rates. Next, we looked in HapMap for any SNPs that lay outside these recombination-determined boundaries with r2 > 0.2 to the focal SNPs in the CEU population and, if necessary, extended the boundaries of the regions to include such SNPs. Finally, for CAD and T2D, we looked in the WTCCC GWAS data for any SNPs with association P value within two orders of magnitude of that of the focal SNP and, if necessary, extended the boundaries to include any such SNPs. In most cases, the boundaries defined by genetic distance did not need to be extended to meet the other criteria.

We attempted to design assays for and genotype all SNPs from the Affymetrix Human 500K SNP array used in our original GWAS study, all polymorphic HapMap SNPs and all SNPs from our resequencing pilot and the other (smaller) resequencing data sets we had access to for the regions we studied, including mining of the Watson and Venter genomes. We did the same for any SNP in dbSNP (version 128) that had genotype or frequency data showing variation and any SNPs that had been reported by more than one group (Table 1).

Genotype calling was performed in two stages. First, genotypes were called using Illuminus9. Only SNPs that Illuminus called with high confidence were taken forward directly; the remainder underwent manual cluster inspection and were called again where appropriate. The first quality control filter applied was to remove individual genotypes with an Illuminus confidence score of less than 0.2, which is the recommended threshold. Samples with call rates lower than 90% were excluded. We excluded SNPs with call rates less than 0.95, Hardy-Weinberg equilibrium P values of less than 0.001 or MAFs less than 0.001.

Imputation

For imputation, we used as a reference panel the 286 haplo-types for individuals of European descent (defined as samples from the CEU, British in England and Scotland (GBR), Iberian population in Spain (IBS) and Toscani in Italia (TSI) populations) from the June 2011 release of the 1000 Genomes Project (see URLs). We used the software package IMPUTE v1.1.5 to perform the imputation. Quality control after imputation consisted of excluding imputed SNPs with either (i) average maximum posterior (as returned by IMPUTE1) less than 0.98, (ii) IMPUTE1 info score less than 0.8 or (iii) greater than 2% missing data. Imputed data were analyzed for association in the program SNPTEST.

ACKNOWLEDGMENTS

We are grateful to D. Altshuler and colleagues at the Broad Institute for provision of SNP discovery information in some of our fine-mapping regions. We acknowledge the many physicians, research fellows and research nurses who contributed to the various case collections and the collection teams and senior management of the UK Blood Services responsible for the UK Blood Services Collection. The principal funder of the project was the Wellcome Trust. For the 1958 Birth Cohort, venous blood collection was funded by the UK Medical Research Council, and cell line production and DNA extraction and processing were funded by the Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory. Many of the authors of this paper received funding from the NIHR Biomedical Research Centers.

Footnotes

URLs. Catalog of Published Genome-Wide Association Studies, http://www.genome.gov/GWAStudies/; 1000 Genomes Project reference panel haplotypes, http://www.1000genomes.org/.

Note: Supplementary information is available in the online version of the paper.

COMPETING FINANCIAL INTERESTS The authors declare no competing financial interests.

Reprints and permissions information is available online at http://www.nature.com/reprints/index.html.

References

  • 1.Steinman L. Mixed results with modulation of TH-17 cells in human autoimmune diseases. Nat. Immunol. 2010;11:41–44. doi: 10.1038/ni.1803. [DOI] [PubMed] [Google Scholar]
  • 2.ENCODE Project Consortium Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447:799–816. doi: 10.1038/nature05874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Stranger BE, et al. Patterns of cis regulatory variation in diverse human populations. PLoS Genet. 2012;8:e1002639. doi: 10.1371/journal.pgen.1002639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Dendrou CA, et al. Cell-specific protein phenotypes for the autoimmune locus IL2RA using a genotype-selectable human bioresource. Nat. Genet. 2009;41:1011–1015. doi: 10.1038/ng.434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.WTCCC Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–678. doi: 10.1038/nature05911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Morris AP, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat. Genet. 2012;44:981–990. doi: 10.1038/ng.2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Peden JF, Farrall M. Thirty-five common variants for coronary artery disease: the fruits of much collaborative labour. Hum. Mol. Genet. 2011;20:R198–R205. doi: 10.1093/hmg/ddr384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Burton PR, et al. Association scan of 14,500 nonsynonymous SNPs in four diseases identifies autoimmunity variants. Nat. Genet. 2007;39:1329–1337. doi: 10.1038/ng.2007.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Teo YY, et al. A genotype calling algorithm for the Illumina BeadArray platform. Bioinformatics. 2007;23:2741–2746. doi: 10.1093/bioinformatics/btm443. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES