#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Bayesian multivariate reanalysis of large genetic studies identifies many new associations


Authors: Michael C. Turchin aff001;  Matthew Stephens aff001
Authors place of work: Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America aff001;  Department of Statistics, The University of Chicago, Chicago, Illinois, United States of America aff002
Published in the journal: Bayesian multivariate reanalysis of large genetic studies identifies many new associations. PLoS Genet 15(10): e32767. doi:10.1371/journal.pgen.1008431
Category: Research Article
doi: https://doi.org/10.1371/journal.pgen.1008431

Summary

Genome-wide association studies (GWAS) have now been conducted for hundreds of phenotypes of relevance to human health. Many such GWAS involve multiple closely-related phenotypes collected on the same samples. However, the vast majority of these GWAS have been analyzed using simple univariate analyses, which consider one phenotype at a time. This is despite the fact that, at least in simulation experiments, multivariate analyses have been shown to be more powerful at detecting associations. Here, we conduct multivariate association analyses on 13 different publicly-available GWAS datasets that involve multiple closely-related phenotypes. These data include large studies of anthropometric traits (GIANT), plasma lipid traits (GlobalLipids), and red blood cell traits (HaemgenRBC). Our analyses identify many new associations (433 in total across the 13 studies), many of which replicate when follow-up samples are available. Overall, our results demonstrate that multivariate analyses can help make more effective use of data from both existing and future GWAS.

Keywords:

Phenotypes – Alleles – Genome-wide association studies – blood pressure – Genetic polymorphism – Open data

Introduction

Genome wide association studies (GWAS) have been widely used to identify genetic factors—particularly single nucleotide polymorphisms (SNPs) and copy number variations (CNVs)—associated with human disease risk and other phenotypes of interest [1, 2]. Indeed, at time of writing over 24,000 such associations have been identified as ‘genome-wide significant’ [3].

The vast majority of these many genetic association analyses consider only one phenotype at a time (“univariate association analysis”). This is despite the fact that measurements on multiple phenotypes are often available, and joint association analysis of multiple phenotypes (“multivariate association analysis”) can substantially increase power [48]. There are likely multiple reasons for the preponderance of univariate analyses. One possible reason is that initial association analyses are usually performed under tight time constraints, and at a time when many other analysis issues (e.g. quality control, population stratification) are competing for attention. In these conditions it makes sense to focus on the simplest possible approach that will quickly yield new associations, without overly worrying about loss of efficiency. In addition analysts may be legitimately concerned that deviation from the most widely adopted analysis pipeline may invite unwanted additional reviewer attention.

Nonetheless, we believe that multivariate association analysis has an important role to play in making the most of costly and time-consuming GWAS studies. One way forward is to conduct multivariate analyses of previously-published GWAS, checking for additional associations that may have been missed by the initial univariate association analyses. This is greatly facilitated by the fact that many GWAS now make summary data from single-SNP tests freely available [913], and that simple multivariate analysis can be conducted using such summary data [1416].

Here we demonstrate the potential benefits of reanalyzing published GWAS using multivariate methods. Specifically we apply multivariate methods from [14] to reanalyze 13 different GWAS whose initial publications reported only univariate results. In most cases our multivariate analyses find many new associations. For example, in GIANT 2014/5 we find over 150 new associations. In studies with multiple data releases, we find that new multivariate associations found in initial releases typically replicate in subsequent releases, supporting that many of the new associations are likely real. We also demonstrate that the multivariate approach is not equivalent to simply relaxing the univariate GWAS significance threshold. Finally, we point out some limitations of the specific framework we used here, and suggest some alternative strategies that may help address those limitations in future multivariate GWAS analyses.

Results

Multivariate association analyses

To facilitate multivariate association analyses using the methods from [14], we implemented them in an R package bmass (Bayesian multivariate analysis of summary statistics). The software requires as input univariate GWAS summary statistics, for the same set of SNPs, on d related phenotypes. (The derivations in [14] are for quantitative phenotypes, but the methods can also be applied to summary data from binary phenotypes, which can be interpreted as making a normal approximate to the likelihood for the effect sizes as in [17].) Then, for each SNP, it attempts to categorize each phenotype as belonging to one of three categories: Unassociated, Directly Associated, or Indirectly Associated with the SNP. The difference between D and I is that an indirect association disappears after controlling for associations with other phenotypes (see Methods and S1 Fig).

For d phenotypes, there are 3d possible assignments of phenotypes to these 3 categories, and each assignment corresponds to a different “model” γ. For example, one model corresponds to the “null” that all phenotypes are Unassociated; another model corresponds to the model that all phenotypes are Directly associated; another model corresponds to just the first phenotype being Directly associated, etc. The goal of the association analysis is to determine which of these models is consistent with the data and, in particular, to assess overall evidence against the null model.

The support in the data for model γ, relative to the null model, is summarized by a Bayes Factor (BFγ). Large values of BFγ indicate strong evidence for model γ compared against the null. One advantage of Bayes Factors over p-values is that the Bayes Factors from different models can be easily compared and combined. For example, the overall evidence against the null is given by the (weighted) average of these BFs:


where the weights wγ are chosen to reflect the relative plausibility of each model γ. In bmass we implemented the Empirical Bayes approach from [14] that learns appropriate weights from the data (see Methods).

Comparisons with published univariate analyses

To provide a benchmark against which to compare our multivariate analysis results, we compiled a list of “previous univariate associations”: SNPs that were both reported as significant in the original publication and exceeded the original publication’s definition for genome-wide significance in at least one phenotype in the publicly-available (univariate) summary data analyzed here. This does not include all SNPs reported in every original publication because in some studies SNPs became genome-wide significant only after adding additional samples not included in the publicly available summary data.

We used these previous univariate associations to determine a significance threshold for our multivariate associations. Specifically, we declared a multivariate association as significant if its BFav exceeds that of any previous univariate association’s BFav in the same study [14]. The rationale is that the evidence for these multivariate associations exceeds the evidence for previously-reported genome-wide significant associations, which are generally regarded as likely to be (mostly) real associations.

Finally, we defined a list of “new multivariate associations”, which are SNPs that are significant in our multivariate analysis but are not a “previous univariate association”. To avoid double-counting of signals due to linkage disequilibrium (LD), we pruned the list of new multivariate associations so that they are all at least 0.5Mb apart. For additional details, see Methods.

Many new loci identified in reanalyzing 13 publicly available GWAS studies

We applied bmass to 13 publicly available GWAS studies, representing 10 different collections of phenotypes (Table 1). Phenotypic collections include blood lipid traits (GlobalLipids: [9, 18]), body morphological traits (GIANT: [1012, 1921]), red blood cell traits (HaemgenRBC: [13, 22]), blood pressure traits [23, 24], bone density traits [25], and kidney function traits [26, 27]. For three of these phenotypic collections (GlobalLipids, GIANT, and HaemgenRBC), two different releases were available from the source consortiums. We conducted basic QC as described in Methods.

Tab. 1. Dataset summary.
Dataset summary.

Our multivariate analyses identify, in total, hundreds of new associations. The numbers of previous univariate associations and new multivariate associations are summarized in Fig 1 (see also Table 2). For example, we identify 162 new multivariate associations in GIANT2014/5, 65 in GlobalLipids2013, and 60 in HaemgenRBC2016. These represent power increases from 10% to 45% compared with previous univariate analyses.

Number of independent significant SNPs, by study.
Fig. 1. Number of independent significant SNPs, by study.
The barplot shows the number of independent SNPs that were significant in previous univariate analyses (blue) and the number of additional significant associations in our new multivariate analyses (red). For univariate analysis, significance levels were set by the original study. For multivariate analyses, we declared a SNP to be significant if its weighted average Bayes Factor (BFav) exceeded that of the smallest BFav among the previous univariate significant SNPs. We considered SNPs more than.5Mb apart to be independent. See Table 1 and Methods for phenotype details, Methods for further analysis details, and S2S4 Tables for lists of significant SNPs from each dataset.
Tab. 2. Summary of new multivariate associations identified.
Summary of new multivariate associations identified.

Replication of multivariate associations across releases

To demonstrate that many of these new multivariate associations are likely to be real we take advantage of three datasets that each have two releases separated by several years (GlobalLipids, GIANT, and HaemgenRBC). In each case we performed multivariate association analysis of the earlier release and checked how the new multivariate associations fared in univariate analyses of the later release (Fig 2). Since later releases include the samples from earlier releases, to assess “replication” we focus on whether the association in the new release is more significant than the original release—that is, whether the signal in the new (non-overlapping) samples provides additional evidence over and above the original signal. By this measure the results show high replication rates for the new multivariate associations: in total, 84 of 94 new associations have smaller minimum univariate p-values in the later release (at exactly the same SNP), and indeed the majority of these reach univariate GWAS significance in the later release.

Replication of new multivariate associations.
Fig. 2. Replication of new multivariate associations.
The figure shows results based on earlier and later releases from studies with multiple releases (GlobalLipids, GIANT, and HaemgenRBC). Each point represents a new multivariate association identified in our multivariate analysis of the earlier release. The x- and y-axes show the minimum (across phenotypes) of the -log10 univariate p-values from the earlier release (x-axis) vs. the later release (y-axis). Dashed red lines represent the univariate significance GWAS thresholds used for each study’s releases. Across all three studies, 84 out of 94 new multivariate associations from the earlier releases have smaller minimum univariate p-values in the later release, and 68 out of 84 new multivariate associations that did not reach GWAS significance in the earlier release do so in the later release (see S5 Table for a per-dataset breakdown).

Multivariate analysis is different from multiple univariate analyses

Because multivariate analysis takes account of joint patterns across phenotypes, its ranking of significance of SNPs can change compared with that from the univariate p-values alone. That is, multivariate analysis is not simply equivalent to multiple univariate analyses. To illustrate this we examined, in three well-powered studies, the associations that would be declared significant if the univariate significance threshold were relaxed, and assessed which of them would also be significant in our multivariate analysis (i.e. we assess whether, if we go deeper into the univariate results, we find the same SNPs as appear in our multivariate results). The results are shown in Fig 3. Although there is, understandably, substantial overlap between the significant SNPs, any non-trivial relaxation of the univariate threshold includes many SNPs that are not multivariate significant in our analysis; for example, at a univariate threshold of 5 × 10−7 only 66% of the univariate significant SNPs are also multivariate significant across these three studies. This demonstrates that, indeed, our multivariate approach reorders significance of SNPs compared with multiple univariate analyses.

Comparison of new multivariate hits vs. relaxing univariate <i>p</i>-value threshold.
Fig. 3. Comparison of new multivariate hits vs. relaxing univariate p-value threshold.
For each data set the graph shows how many associations become significant as the univariate p-value threshold is relaxed (moving from right to left on the x-axis), and how many of these are declared as new multivariate hits in our analysis. In both cases results are pruned to avoid counting associations of SNPs in strong LD; see Methods for details. The appearance of appreciable blue areas indicates that the multivariate analysis is reordering the significance of SNPs compared with performing multiple univariate analyses.

Reanalysis also identifies new univariate associations

During our multivariate reanalyses we noticed many SNPs that appeared to be genome-wide univariate significant but were—somewhat mysteriously—not reported as such by the original studies (i.e. SNPs whose univariate p-values crossed the significance threshold, as defined by the given study, in at least one trait). S1 Table reports 79 such associations.

There may be many reasons why such variants went unreported, but one reason may be physical proximity to a variant with a stronger signal. Indeed, more than half of the variants described above are within 1Mb of a previously-reported univariate GWAS association. Refraining from reporting multiple near-by associations seems a reasonable—if conservative—strategy to avoid reporting redundant associations due to LD. Further, even when redundant associations due to LD can be ruled out (e.g. by directly examining LD rather than by simply using physical distance), it might be assumed that multiple nearby associated variants may all act through the same biological mechanism and therefore provide redundant biological insights. However, we found that multi-phenotype patterns of association can differ between nearby SNPs, suggesting that they act through different mechanisms.

To highlight just one example, consider rs7515577—which is an original univariate association in GlobalLipids2010—and rs12038699—which is a new multivariate association in GlobalLipids2013. We note that rs12038699 actually reached univariate genome-wide significance in the GlobalLipids2013 dataset, but was not reported (S6 Table). This is possibly because the latter SNP is relatively close, in genomic terms, to the former SNP (549kb). However, these SNPs are not in strong LD (r2 = .08), and so these associations almost certainly represent non-redundant associations. This is further supported by the effect sizes in each phenotype, which clearly reveal very different multivariate patterns of effect sizes among phenotypes (S2 Fig & S6 Table). Indeed the very different multivariate patterns of effect size suggest that not only are these associations non-redundant but likely involve different biological mechanisms as well.

These results suggest that, moving forward, it may pay to be more careful in designing filters designed to avoid reporting redundant associations, and that multi-phenotype analyses may have a helpful role to play here.

Limitations

One goal of the multivariate approach introduced in [14] was to increase interpretability of multivariate analyses; in particular, the goal was to not only test for associations but also to help explain the associations by partitioning the phenotypes into “Unassociated”, “Directly Associated”, and “Indirectly Associated” categories. In principle one might hope to use these classifications to gain insights into the relationships among phenotypes and also perhaps to identify different “types” of multivariate association—effectively clustering associations into different groups. However, in practice we find that these discrete classifications are often not as helpful as one might hope. One reason is the difficulty of reliably distinguishing between direct and indirect effects [14]. Another reason is widespread associations with multiple phenotypes. Indeed, we find that, consistently across data sets, the most common multivariate models involve associations—either direct or indirect—with many phenotypes (S7 Table) and many SNPs are classified as being associated with many phenotypes (Fig 4A). Further, SNPs are very rarely confidently classified as “Unassociated” with any phenotype (Fig 4B). This last observation can be explained by the fact that it is essentially impossible to distinguish ‘unassociated’ from ‘weakly associated’. Nonetheless when all SNPs show similar classifications it is difficult to get insights into different patterns of multivariate association.

Distribution, across significant SNPs, of number of phenotypes that are confidently associated (A) or confidently unassociated (B).
Fig. 4. Distribution, across significant SNPs, of number of phenotypes that are confidently associated (A) or confidently unassociated (B).
Results are shown for three well-powered datasets: GlobalLipids2013, GIANT2014/5, and HaemgenRBC2016. Here “confident” means with probability >0.95, so a SNP is considered “confidently associated” with a phenotype if the sum of its probabilities in the “Directly Associated” and “Indirectly Associated” categories exceeds 0.95 (A), and is considered confidently unassociated with the phenotype if this probability is less than 0.05 (B). The set of significant SNPs includes both previous univariate associations and new multivariate associations.

Moving forward, rather than relying on the discrete classifications of “Unassociated”, “Directly Associated”, and “Indirectly Associated” to identify different patterns of multivariate association, we believe it will be more fruitful to use multivariate methods that take a more quantitative approach, such as identifying different patterns of effect size (including direction of effect) among phenotypes [28]. Focusing on effect sizes has the potential to be much more informative than discrete classification, which can hide effect size differences. For example, when multiple SNPs are classified as associated with all phenotypes, they can still show very different patterns of estimated effect sizes/direction (see S3 Fig).

Another limitation of our multivariate methods is that they can lead to (what appear to be) false positive associations when applied to test SNPs with very low minor allele frequencies. Specifically we saw examples where very low-frequency SNPs (e.g. MAF < .001) showed strong signals of multivariate association despite showing very little signal in any univariate test. Although such results are not impossible, we believe that most of these cases were likely false positives, and we applied a MAF cut-off (of 0.01 or 0.005) to avoid these issues. Consequently we recommend caution in interpreting results of multivariate analyses at very low-frequency SNPs, and more generally we recommend that multivariate results be compared against univariate results to check they make sense—highly significant multivariate associations that do not also show at least a moderate level of univariate association should be treated with caution.

Discussion

We reanalyzed 13 publicly available GWAS datasets using a Bayesian multivariate approach and identified many new genetic associations. Turning genetic associations into biological discoveries remains, of course, a challenging problem. Nonetheless, our results suggest that the increased power of multivariate association analysis that has been reported in many simulation studies [8, 14, 29] also translates to discovery of many new associations in practice.

Our results exploit the public availability of summary data from several large GWAS. Despite progress toward easier availability of individual-level data for large studies [30], in many cases summary data remain much easier to obtain and work with; there are big practical advantages as well to modular pipelines that first compute summary data and then use these as inputs to subsequent (more sophisticated) analyses. For example, the multivariate analyses we present here are simplified by assuming that the summary data were computed while adequately adjusting for population stratification and other relevant covariates (indeed, our current bmass software implementation does not allow adjustment for covariates, and so any adjustment must be done in the univariate analyses). And our results illustrate the potential for reanalysis of summary data to yield novel inferences. In this regard we also emphasize the importance of consortia releasing carefully-chosen summaries. For example, Z-scores are much more helpful than p-values because they preserve information on the direction of the effect. Even better would be both the effect size and standard error that created the Z-score. More generally, it is always helpful to include additional key meta-data (e.g. the reference allele, or effect allele, the minor allele frequency, and sample size).

The specific multivariate methods used here were derived under the assumption that the summary data from each phenotype has been obtained from the same sampled individuals (which is true, at least approximately, for studies analyzed here). However, multivariate analysis of summary data is also possible even when data were obtained from different samples for each phenotype. The main difference between these settings is that, for data from overlapping samples, the “noise” is correlated as well as the signal: i.e. the summary data are correlated under the null due to sample overlap, and correlated under the alternative due to both sample overlap and any shared genetic effects. In contrast, for data from non-overlapping samples the noise is uncorrelated (whereas the signal may remain correlated if genetic factors are shared). Our methods use data at (empirically) null SNPs to estimate the noise correlation, and so their overall assessment of associations should be relatively robust to whether samples for different phenotypes overlap (however, our definitions of D (direct) vs I (indirect) associations requires the same samples to be measured across phenotypes.)

Moving forward, we expect multivariate association analyses to play an increasingly important role in detecting and understanding genetic associations and relationships among phenotypes. Large studies are now collecting, and making available, rich human genetic and phenotypic information on many complex phenotypes, most notably the UKBioBank [30]. In addition, there are increasingly large studies linking genetic variation and molecular phenotypes such as gene expression (e.g. the GTEx project [31]), as well as epigenetic modifications and transcript degradation [3235]. Analysis of multiple molecular traits can help yield insights into causal connections among traits [36], and joint analysis of molecular traits with complex phenotypes may also shed light on functional mechanisms (as in “co-localization” analyses [16, 3739]). Even simply moving from single phenotype to pairwise analysis can shed considerable light on sharing of genetic effects and possible causal connections [15, 40].

These increasingly-complex new data also bring new analytic and computational challenges. Here we have restricted our analyses to relatively small sets of closely-related traits, and indeed the specific multivariate framework we used here—which performs an exhaustive search over all possible multivariate models—is fully tractable for only moderate numbers of traits (up to about 10). Scaling methods up to dealing with larger number of traits may well be helpful for some settings, and recent multivariate analysis methods can deal with dozens of outcomes [28, 41]. In addition, developing multivariate methods to perform fine-mapping of genetic associations simultaneously across multiple phenotypes [42] seems an important and challenging area for future work.

URLs

bmass R package: https://github.com/mturchin20/bmass.

Methods

GWAS datasets

Below are specific details regarding retrieval and data-processing for each dataset analyzed. Where applicable, these details include the sample size (N), minor allele frequency (MAF), and p-value thresholds that were applied (based on the thresholds used in the original publications). For each dataset variants were dropped if they satisfied at least one of the following criteria: did not contain information for every phenotype; had missing MAF; were fixed (MAF of 0); had effect size exactly 0 (i.e. direction of effect would be indeterminable); or did not contain the same reference and alternative alleles across each phenotype. For a handful of studies, external databases were used to retrieve chromosome, basepair information, and MAF based on rsID#; in these studies SNPs for which this information could not be retrieved were also dropped.

GlobalLipids2010 [18]: Original merged, processed, and GWAS-hit annotated summary data from [14] for HDL, LDL, TG, and TC was downloaded from https://github.com/stephens999/multivariate (dtlesssignif.annot.txt and RSS0.txt).

GlobalLipids2013 [9]: Summary data for HDL, LDL, TG, and TC was downloaded from http://csg.sph.umich.edu/abecasis/public/lipids2013/. We used a minimum N threshold of 50,000, a MAF threshold of 1%, and a univariate significant GWAS p-value threshold of 5 × 10−8. All variants were oriented to the HDL minor allele. The final merged and QC’d datafile contained 2,004,701 SNPs. rsID#’s of published GWAS SNPs were retrieved for all four phenotypes from https://www.nature.com/ng/journal/v45/n11/full/ng.2797.html via Supplementary Tables 2 and 3.

GIANT2010 [1921]: Summary data for Height, BMI, and WHRadjBMI were downloaded from https://www.broadinstitute.org/collaboration/giant/index.php/GIANT_consortium_data_files. We used a minimum N threshold of 50,000, a MAF threshold of 1%, and a univariate significant GWAS p-value threshold of 5 × 10−8. Chromosome and basepair position per variant were retrieved from dbSNP130 [43]. All variants were oriented to the Height minor allele. The final merged and QC’ed datafile contained 2,363,881 SNPs. rsID#’s of published GWAS SNPs were retrieved for Height from https://www.nature.com/nature/journal/v467/n7317/full/nature09410.html via Supplementary Table 1, for BMI from https://www.nature.com/ng/journal/v42/n11/full/ng.686.html via Table 1, and for WHRadjBMI from https://www.nature.com/ng/journal/v42/n11/full/ng.685.html via Table 1.

GIANT2014/5 [1012]: Summary data for Height, BMI, and WHRadjBMI were downloaded from https://www.broadinstitute.org/collaboration/giant/index.php/GIANT_consortium_data_files. We used a minimum N threshold of 50,000, a MAF threshold of 1%, and a univariate significant GWAS p-value threshold of 5 × 10−8. Chromosome and basepair position per variant were retrieved from dbSNP130 [43]. All variants were oriented to the Height minor allele. The final merged and QC’ed datafile contained 2,340,715 SNPs. rsID#’s of published GWAS SNPs were retrieved for Height from https://www.nature.com/ng/journal/v46/n11/full/ng.3097.html via Supplementary Table 1, for BMI from https://www.nature.com/nature/journal/v518/n7538/full/nature14177.html via Supplementary Tables 1 and 2, and for WHRadjBMI from https://www.nature.com/nature/journal/v518/n7538/full/nature14132.html via Supplementary Table 4.

HaemgenRBC2012 [22]: Summary data for RBC, PCV, MCV, MCH, MCHC, and Hb were downloaded from the European Genome-Phenome Archive via accession number EGAS00000000132 (https://www.ebi.ac.uk/ega/studies/EGAS00000000132). We used a minimum N threshold of 10,000, a MAF threshold of 1%, and a univariate significant GWAS p-value threshold of 1 × 10−8. Chromosome, basepair position, and MAF per variant were retrieved from HapMap release 22 [44]. All variants were oriented to the RBC minor allele. The final merged and QC’ed datafile contained 2,327,567 SNPs. rsID#’s of published GWAS SNPs were retrieved for all six phenotypes from https://www.nature.com/nature/journal/v492/n7429/full/nature11677.html via Table 1.

HaemgenRBC2016 [13]: Summary data for RBC, PCV, MCV, MCH, MCHC, and Hb were shared via personal communication with the authors. We used a MAF threshold of 1% and a univariate significant GWAS p-value threshold of 8.319×10−9. Since sample size was not provided per variant, the following overall study sample sizes were used as proxies per phenotype: 172,952 for RBC, 172,433 for PCV, 173,039 for MCV, 172,332 for MCH, for 172,925 MCHC, and 172,851 for Hb. All variants were oriented to the RBC minor allele. Only SNPs were analyzed. The final merged and QC’ed datafile contained 8,649,095 SNPs. We then used these summary data to create a list of (non-redundant) “Previous univariate associations”. This was done separately for each phenotype by collecting all SNPs that exceeded the univariate significant GWAS p-value threshold and greedily pruning the SNPs: i.e. we went down the list, removing SNPs that were less significant than another SNP within 500kb. The pruned lists of previous univariate associations for each phenotype were then combined to produce the final SNP list of “published GWAS results”. Published CNVs that tagged regions that were not identified by this ‘final published SNP list’ were also included to avoid erroneously claiming downstream a region as a ‘new unpublished result’; these CNVs however were only used to mask additional loci as being ‘nearby a published univariate GWAS result’ and for nothing else in the bmass analysis pipeline.

ICBP2011 [23, 24]: Summary data for SBP, DBP, PP, and MAP were downloaded from dbGaP via accession number phs000585.v1.p1 (https://www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs000585.v1.p1). We used a minimum N threshold of 10,000, a MAF threshold of 1%, and a univariate significant GWAS p-value threshold of 5 × 10−8. Chromosome and basepair position per variant were retrieved from HapMap release 21 [44]. All variants were oriented to the SBP minor allele. The final merged and QC’ed datafile contained 2,387,851 SNPs. rsID#’s of published GWAS SNPs were retrieved for SBP and DBP from https://www.nature.com/nature/journal/v478/n7367/full/nature10405.html via Supplementary Table 5, and for PP and MAP from https://www.nature.com/ng/journal/v43/n10/full/ng.922.html via Table 1 and Supplementary Table 2F. Additionally, we gratefully acknowledge the International Consortium for Blood Pressure Genome-Wide Association Studies (Nature. 2011 Sep 11;478(7367):103-9, Nat Genet. 2011 Sep 11;43(10):1005-11) for generating and sharing these data.

MAGIC2010 [45]: Summary data for FstIns, FstGlu, HOMA_B, and HOMA_IR were downloaded from https://www.magicinvestigators.org/downloads/. We used a MAF threshold of 1% and a univariate significant GWAS p-value threshold of 5 × 10−8. Since sample size was not provided per variant, the overall study sample size of 46,186 was used as a proxy. Chromosome and basepair position per variant were retrieved from HapMap release 22 [44]. All variants were oriented to the FstIns minor allele. The final merged and QC’ed datafile contained 2,333,328 SNPs. rsID#’s of published GWAS SNPs were retrieved for all four phenotypes from https://www.nature.com/ng/journal/v42/n2/full/ng.520.html via Table 1.

GEFOS2015 [25]: Summary data for FA, FN, and LS were downloaded from http://www.gefos.org/?q=content/data-release-2015. We used a MAF threshold of.5% and a univariate significant GWAS p-value threshold of 1.2 × 10−8. Since sample size was not provided per variant, the overall study sample size of 32,965 was used as a proxy. All variants were oriented to the FA minor allele. The final merged and QC’ed datafile contained 8,938,035 SNPs. rsID#’s of published GWAS SNPs were retrieved for all four phenotypes from https://www.nature.com/nature/journal/v526/n7571/full/nature14878.html via Supplementary Table 13.

GIS2014 [46]: Summary data for Iron, Sat, TrnsFrn, and Log10Frtn were shared via personal communication with the authors. We used a MAF threshold of 1% and a univariate significant GWAS p-value threshold of 5 × 10−8. Since sample size was not provided per variant, the overall study sample size of 48,972 was used as a proxy. All variants were oriented to the Iron minor allele. The final merged and QC’ed datafile contained 1,985,313 SNPs. rsID#’s of published GWAS SNPs were retrieved for all four phenotypes from https://www.nature.com/articles/ncomms5926/ via Table 1.

SSGAC2016 [47]: Summary data for NEB_Pooled and AFB_Pooled were downloaded from https://www.thessgac.org/data. We used a MAF threshold of 1% and a univariate significant GWAS p-value threshold of 5 × 10−8. Since sample size was not provided per variant, the following overall study sample sizes were used as proxies per phenotype: 251,151 for NEB_Pooled and 343,072 for AFB_Pooled. All variants were oriented to the NEB_Pooled minor allele. The final merged and QC’ed datafile contained 2,395,561 SNPs. rsID#’s of published GWAS SNPs were retrieved for all four phenotypes from https://www.nature.com/ng/journal/v48/n12/full/ng.3698.html via Table 1.

CKDGen2010/1 [26, 27]: Summary data for Crea, Cys, CKD, UACR, and MA were downloaded from https://www.nhlbi.nih.gov/research/intramural/researchers/pi/fox-caroline/datasets. We used a MAF threshold of 1% and a univariate significant GWAS p-value threshold of 5 × 10−8. Since sample size was not provided per variant, the following overall study sample sizes were used as proxies per phenotype: 67,093 for Crea, 20,957 for Cys, 62,237 for CKD, 31,580 for UACR, and 30,482 for MA. All variants were oriented to the Crea minor allele. The final merged and QC’ed datafile contained 2,333,498 SNPs. rsID#’s of published GWAS SNPs were retrieved for Crea, Cys, and CKD from https://www.nature.com/ng/journal/v42/n5/full/ng.568.html via Table 2.

ENIGMA22015 [48]: Summary data for ICV, Accumbens, Amygdala, Caudate, Hippocampus, Pallidum, Putamen, and Thalamus were downloaded from http://enigma.ini.usc.edu/research/download-enigma-gwas-results/. We used a minimum N threshold of 10,000, a MAF threshold of 1% and a univariate significant GWAS p-value threshold of 5 × 10−8. All variants were oriented to the ICV minor allele. The final merged and QC’ed datafile contained 6,271,117 SNPs. rsID#’s of published GWAS SNPs were retrieved for all 8 phenotypes from https://www.nature.com/nature/journal/v520/n7546/full/nature14101.html via Table 1.

bmass

bmass implements in an R package the statistical methods described in [14], which should be consulted for full details. In particular, the sections “Computation” and “Detailed Methods (Global Lipids Analysis)” in [14] describe how multivariate analyses are applied to GWAS summary data, and bmass implements the data analysis pipeline described in the “Detailed Methods (Global Lipids Analysis)” section. The bmass R package also includes two vignettes to help users begin to process GWAS summary data and implement these methods.

Additional details for Fig 3

For each dataset we made a list of “marginally-significant” SNPs, with p-values smaller than 1 × 10−6 but not genome-wide significant at the relevant datasets’ GWAS threshold. We then greedily pruned these lists of marginally-significant SNPs: that is we repeatedly went through the lists removing SNPs that were less significant than another SNP within 500kb. We then removed any SNPs that were within 500kb of a new multivariate association, and merged the resulting list with the list of new multivariate associations, and sorted this merged list of SNPs by their minimum univariate p-value.

This results in a non-redundant list of marginally-significant SNPs—some of which are new multivariate associations and some of which are not—sorted by their smallest univariate p-value. The plot shows how the number of SNPs of each type varies as the p-value threshold is relaxed from the GWAS threshold to 10−6 (the HaemgenRBC2016 results show only the top 500 SNPs due to the abundance of SNPs between 8.31 × 10−9 and 1 × 10−6).

Supporting information

S1 Fig [dag]
Graphical model of multivariate categories.

S2 Fig [pdf]
Refining association signals—GlobalLipids2013 rs7515577 and rs12038699.

S3 Fig [bars]
Effect size heterogeneity among SNPs with identical multivariate model assignments.

S1 Table [pdf]
Summary of associations in each dataset.

S2 Table [xls]
Lists of new multivariate associations, per dataset.

S3 Table [s]
Lists of retrieved univariate associations from original publications, per dataset.

S4 Table [xls]
Results for previous univariate associations, per dataset.

S5 Table [bmass]
Replication of new multivariate associations.

S6 Table [tc]
-Values for rs7515577 & rs12038699 in 2010 and 2013 GlobalLipids releases.

S7 Table [pdf]
Top multivariate model examples per SNP.


Zdroje

1. Price AL, Spencer CC, Donnelly P. Progress and promise in understanding the genetic basis of common diseases. Proc Biol Sci. 2015;282(1821):20151684. doi: 10.1098/rspb.2015.1684 26702037

2. Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA, et al. 10 Years of GWAS Discovery: Biology, Function, and Translation. Am J Hum Genet. 2017;101(1):5–22. doi: 10.1016/j.ajhg.2017.06.005 28686856

3. MacArthur J, Bowler E, Cerezo M, Gil L, Hall P, Hastings E, et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 2017;45(D1):D896–D901. doi: 10.1093/nar/gkw1133 27899670

4. Jiang C, Zeng ZB. Multiple trait analysis of genetic mapping for quantitative trait loci. Genetics. 1995;140(3):1111–27. 7672582

5. Zhu W, Zhang H. Why Do We Test Multiple Traits in Genetic Association Studies? J Korean Stat Soc. 2009;38(1):1–10. doi: 10.1016/j.jkss.2008.10.006 19655045

6. Shriner D. Moving toward System Genetics through Multiple Trait Analysis in Genome-Wide Association Studies. Front Genet. 2012;3:1. doi: 10.3389/fgene.2012.00001 22303408

7. Yang Q, Wang Y. Methods for Analyzing Multivariate Phenotypes in Genetic Association Studies. J Probab Stat. 2012;2012:652569. doi: 10.1155/2012/652569 24748889

8. Galesloot TE, van Steen K, Kiemeney LA, Janss LL, Vermeulen SH. A comparison of multivariate genome-wide association methods. PLoS One. 2014;9(4):e95923. doi: 10.1371/journal.pone.0095923 24763738

9. Willer CJ, Schmidt EM, Sengupta S, Peloso GM, Gustafsson S, Kanoni S, et al. Discovery and refinement of loci associated with lipid levels. Nat Genet. 2013;45(11):1274–1283. doi: 10.1038/ng.2797 24097068

10. Wood AR, Esko T, Yang J, Vedantam S, Pers TH, Gustafsson S, et al. Defining the role of common variation in the genomic and biological architecture of adult human height. Nat Genet. 2014;46(11):1173–86. doi: 10.1038/ng.3097 25282103

11. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, et al. Genetic studies of body mass index yield new insights for obesity biology. Nature. 2015;518(7538):197–206. doi: 10.1038/nature14177 25673413

12. Shungin D, Winkler TW, Croteau-Chonka DC, Ferreira T, Locke AE, Magi R, et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature. 2015;518(7538):187–196. doi: 10.1038/nature14132 25673412

13. Astle WJ, Elding H, Jiang T, Allen D, Ruklisa D, Mann AL, et al. The Allelic Landscape of Human Blood Cell Trait Variation and Links to Common Complex Disease. Cell. 2016;167(5):1415–1429 e19. doi: 10.1016/j.cell.2016.10.042 27863252

14. Stephens M. A unified framework for association analysis with multiple related phenotypes. PLoS One. 2013;8(7):e65245. doi: 10.1371/journal.pone.0065245 23861737

15. Pickrell JK, Berisa T, Liu JZ, Segurel L, Tung JY, Hinds DA. Detection and interpretation of shared genetic influences on 42 human traits. Nat Genet. 2016;48(7):709–17. doi: 10.1038/ng.3570 27182965

16. Hormozdiari F, van de Bunt M, Segre AV, Li X, Joo JWJ, Bilow M, et al. Colocalization of GWAS and eQTL Signals Detects Target Genes. Am J Hum Genet. 2016;99(6):1245–1260. doi: 10.1016/j.ajhg.2016.10.003 27866706

17. Wakefield J. Bayes factors for genome-wide association studies: comparison with P-values. Genet Epidemiol. 2009;33(1):79–86. doi: 10.1002/gepi.20359 18642345

18. Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M, et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature. 2010;466(7307):707–13. doi: 10.1038/nature09270 20686565

19. Lango Allen H, Estrada K, Lettre G, Berndt SI, Weedon MN, Rivadeneira F, et al. Hundreds of variants clustered in genomic loci and biological pathways affect human height. Nature. 2010;467(7317):832–8. doi: 10.1038/nature09410 20881960

20. Speliotes EK, Willer CJ, Berndt SI, Monda KL, Thorleifsson G, Jackson AU, et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat Genet. 2010;42(11):937–48. doi: 10.1038/ng.686 20935630

21. Heid IM, Jackson AU, Randall JC, Winkler TW, Qi L, Steinthorsdottir V, et al. Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nat Genet. 2010;42(11):949–60. doi: 10.1038/ng.685 20935629

22. van der Harst P, Zhang W, Mateo Leach I, Rendon A, Verweij N, Sehmi J, et al. Seventy-five genetic loci influencing the human red blood cell. Nature. 2012;492(7429):369–75. doi: 10.1038/nature11677 23222517

23. International Consortium for Blood Pressure Genome-Wide Association S, Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, et al. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 2011;478(7367):103–9. doi: 10.1038/nature10405

24. Wain LV, Verwoert GC, O’Reilly PF, Shi G, Johnson T, Johnson AD, et al. Genome-wide association study identifies six new loci influencing pulse pressure and mean arterial pressure. Nat Genet. 2011;43(10):1005–11. doi: 10.1038/ng.922 21909110

25. Zheng HF, Forgetta V, Hsu YH, Estrada K, Rosello-Diez A, Leo PJ, et al. Whole-genome sequencing identifies EN1 as a determinant of bone density and fracture. Nature. 2015;526(7571):112–7. doi: 10.1038/nature14878 26367794

26. Kottgen A, Pattaro C, Boger CA, Fuchsberger C, Olden M, Glazer NL, et al. New loci associated with kidney function and chronic kidney disease. Nat Genet. 2010;42(5):376–84. doi: 10.1038/ng.568 20383146

27. Boger CA, Chen MH, Tin A, Olden M, Kottgen A, de Boer IH, et al. CUBN is a gene locus for albuminuria. J Am Soc Nephrol. 2011;22(3):555–70. doi: 10.1681/ASN.2010060598 21355061

28. Urbut SM, Wang G, Carbonetto P, Stephens M. Flexible statistical methods for estimating and testing effects in genomic studies with multiple conditions. Nat Genet. 2019;51(1):187–195. doi: 10.1038/s41588-018-0268-8 30478440

29. Porter HF, O’Reilly PF. Multivariate simulation framework reveals performance of multi-trait GWAS methods. Sci Rep. 2017;7:38837. doi: 10.1038/srep38837 28287610

30. Sudlow C, Gallacher J, Allen N, Beral V, Burton P, Danesh J, et al. UK biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015;12(3):e1001779. doi: 10.1371/journal.pmed.1001779 25826379

31. GTEx Consortium T. The Genotype-Tissue Expression (GTEx) project. Nat Genet. 2013;45(6):580–5. doi: 10.1038/ng.2653

32. Gaffney DJ. Global properties and functional complexity of human gene regulatory variation. PLoS Genet. 2013;9(5):e1003501. doi: 10.1371/journal.pgen.1003501 23737752

33. Pai AA, Pritchard JK, Gilad Y. The genetic and mechanistic basis for variation in gene regulation. PLoS Genet. 2015;11(1):e1004857. doi: 10.1371/journal.pgen.1004857 25569255

34. Birney E, Smith GD, Greally JM. Epigenome-wide Association Studies and the Interpretation of Disease -Omics. PLoS Genet. 2016;12(6):e1006105. doi: 10.1371/journal.pgen.1006105 27336614

35. Stricker SH, Koferle A, Beck S. From profiles to function in epigenomics. Nat Rev Genet. 2017;18(1):51–66. doi: 10.1038/nrg.2016.138 27867193

36. Li YI, van de Geijn B, Raj A, Knowles DA, Petti AA, Golan D, et al. RNA splicing is a primary link between genetic variation and disease. Science. 2016;352(6285):600–4. doi: 10.1126/science.aad9417 27126046

37. Li Y, Kellis M. Joint Bayesian inference of risk variants and tissue-specific epigenomic enrichments across multiple complex human diseases. Nucleic Acids Res. 2016;44(18):e144. doi: 10.1093/nar/gkw627 27407109

38. Zhu Z, Zhang F, Hu H, Bakshi A, Robinson MR, Powell JE, et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat Genet. 2016;48(5):481–7. doi: 10.1038/ng.3538 27019110

39. Wen X, Pique-Regi R, Luca F. Integrating molecular QTL data into genome-wide genetic association analysis: Probabilistic assessment of enrichment and colocalization. PLoS Genet. 2017;13(3):e1006646. doi: 10.1371/journal.pgen.1006646 28278150

40. Shi H, Mancuso N, Spendlove S, Pasaniuc B. Local Genetic Correlation Gives Insights into the Shared Genetic Architecture of Complex Traits. Am J Hum Genet. 2017;101(5):737–751. doi: 10.1016/j.ajhg.2017.09.022 29100087

41. Dahl A, Iotchkova V, Baud A, Johansson A, Gyllensten U, Soranzo N, et al. A multiple-phenotype imputation method for genetic studies. Nat Genet. 2016;48(4):466–72. doi: 10.1038/ng.3513 26901065

42. Lewin A, Saadi H, Peters JE, Moreno-Moral A, Lee JC, Smith KG, et al. MT-HESS: an efficient Bayesian approach for simultaneous association detection in OMICS datasets, with application to eQTL mapping in multiple tissues. Bioinformatics. 2016;32(4):523–32. doi: 10.1093/bioinformatics/btv568 26504141

43. Sherry ST, Ward MH, Kholodov M, Baker J, Phan L, Smigielski EM, et al. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 2001;29(1):308–11. doi: 10.1093/nar/29.1.308 11125122

44. International HapMap C. The International HapMap Project. Nature. 2003;426(6968):789–96. doi: 10.1038/nature02168

45. Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson AU, et al. New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet. 2010;42(2):105–16. doi: 10.1038/ng.520 20081858

46. Benyamin B, Esko T, Ried JS, Radhakrishnan A, Vermeulen SH, Traglia M, et al. Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis. Nat Commun. 2014;5:4926. doi: 10.1038/ncomms5926 25352340

47. Barban N, Jansen R, de Vlaming R, Vaez A, Mandemakers JJ, Tropf FC, et al. Genome-wide analysis identifies 12 loci influencing human reproductive behavior. Nat Genet. 2016;48(12):1462–1472. doi: 10.1038/ng.3698 27798627

48. Hibar DP, Stein JL, Renteria ME, Arias-Vasquez A, Desrivieres S, Jahanshad N, et al. Common genetic variants influence human subcortical brain structures. Nature. 2015;520(7546):224–9. doi: 10.1038/nature14101 25607358

49. Genomes Project C, Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68–74. doi: 10.1038/nature15393

Štítky
Genetika Reprodukční medicína

Článek vyšel v časopise

PLOS Genetics


2019 Číslo 10

Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#