Skip to main content
Elsevier Sponsored Documents logoLink to Elsevier Sponsored Documents
. 2021 Sep;75(3):572–581. doi: 10.1016/j.jhep.2021.04.055

An international genome-wide meta-analysis of primary biliary cholangitis: Novel risk loci and candidate drugs

Heather J Cordell 1, James J Fryett 1, Kazuko Ueno 2, Rebecca Darlay 1, Yoshihiro Aiba 3, Yuki Hitomi 4, Minae Kawashima 4, Nao Nishida 4, Seik-Soon Khor 2, Olivier Gervais 5,6, Yosuke Kawai 2, Masao Nagasaki 5,6, Katsushi Tokunaga 2, Ruqi Tang 7, Yongyong Shi 8,9, Zhiqiang Li 8,9, Brian D Juran 10, Elizabeth J Atkinson 11, Alessio Gerussi 12,13, Marco Carbone 12,13, Rosanna Asselta 14,15, Angela Cheung 10, Mariza de Andrade 11, Aris Baras 16, Julie Horowitz 16, Manuel AR Ferreira 16, Dylan Sun 16, David E Jones 17, Steven Flack 18, Ann Spicer 18, Victoria L Mulcahy 18, Jinyoung Byan 19, Younghun Han 19, Richard N Sandford 18, Konstantinos N Lazaridis 10, Christopher I Amos 19, Gideon M Hirschfield 20, Michael F Seldin 21, Pietro Invernizzi 12,13, Katherine A Siminovitch 22,23, Xiong Ma 7, Minoru Nakamura 3,24, George F Mells 18,; Canadian PBC Consortium; Chinese PBC Consortium; Italian PBC Study Group; Japan-PBC-GWAS Consortium; US PBC Consortium; UK-PBC Consortium, for the PBC Consortia; for the ; for the ; for the ; for the ; for the ; for the
PMCID: PMC8811537  PMID: 34033851

Abstract

Backgrounds & Aims

Primary biliary cholangitis (PBC) is a chronic liver disease in which autoimmune destruction of the small intrahepatic bile ducts eventually leads to cirrhosis. Many patients have inadequate response to licensed medications, motivating the search for novel therapies. Previous genome-wide association studies (GWAS) and meta-analyses (GWMA) of PBC have identified numerous risk loci for this condition, providing insight into its aetiology. We undertook the largest GWMA of PBC to date, aiming to identify additional risk loci and prioritise candidate genes for in silico drug efficacy screening.

Methods

We combined new and existing genotype data for 10,516 cases and 20,772 controls from 5 European and 2 East Asian cohorts.

Results

We identified 56 genome-wide significant loci (20 novel) including 46 in European, 13 in Asian, and 41 in combined cohorts; and a 57th genome-wide significant locus (also novel) in conditional analysis of the European cohorts. Candidate genes at newly identified loci include FCRL3, INAVA, PRDM1, IRF7, CCR6, CD226, and IL12RB1, which each play key roles in immunity. Pathway analysis reiterated the likely importance of pattern recognition receptor and TNF signalling, JAK-STAT signalling, and differentiation of T helper (TH)1 and TH17 cells in the pathogenesis of this disease. Drug efficacy screening identified several medications predicted to be therapeutic in PBC, some of which are well-established in the treatment of other autoimmune disorders.

Conclusions

This study has identified additional risk loci for PBC, provided a hierarchy of agents that could be trialled in this condition, and emphasised the value of genetic and genomic approaches to drug discovery in complex disorders.

Lay summary

Primary biliary cholangitis (PBC) is a chronic liver disease that eventually leads to cirrhosis. In this study, we analysed genetic information from 10,516 people with PBC and 20,772 healthy individuals recruited in Canada, China, Italy, Japan, the UK, or the USA. We identified several genetic regions associated with PBC. Each of these regions contains several genes. For each region, we used diverse sources of evidence to help us choose the gene most likely to be involved in causing PBC. We used these ‘candidate genes’ to help us identify medications that are currently used for treatment of other conditions, which might also be useful for treatment of PBC.

Keywords: UK-PBC, ERN RARE-LIVER, ALSPAC, Genomic co-localization, Network-based in silico drug efficacy screening

Graphical abstract

graphic file with name ga1.jpg

Highlights

  • Trans-ethnic genome-wide meta-analysis (GWMA) of susceptibility to primary biliary cholangitis (PBC).

  • Five cohorts of European ancestry and two East Asian cohorts (n = 10,516 cases and 20,772 controls).

  • Identification of 21 additional risk loci for PBC.

  • Preliminary evidence that the genetic architecture of PBC is broadly shared across European and East Asian populations.

  • Identification (using in silico drug efficacy screening) of medications potentially suitable for re-purposing to PBC.

Introduction

Primary biliary cholangitis (PBC) is a chronic liver disease in which autoimmune injury to the small intrahepatic bile ducts eventually leads to cirrhosis. Only 2 medications, ursodeoxycholic acid (UDCA) and obeticholic acid (OCA), are licensed for the treatment of PBC. Many patients have inadequate response to both agents, leaving them at risk of progressive liver disease. Notwithstanding recent advances, novel therapies are needed for this condition.

Delineating the genetic architecture of PBC can provide insight into its aetiology – and more specifically, identify potential drug targets. Therefore, over the past decade, our respective groups have undertaken genome-wide association studies (GWAS) of PBC in Canadian-US,1 Italian,2 British,3 Japanese,4 and Chinese5 cohorts; and in 2015, we undertook a genome-wide meta-analysis (GWMA) of the Canadian-US, Italian, and British discovery panels.6 These studies have identified genome-wide significant associations at the human leukocyte antigen (HLA) locus and 42 non-HLA loci.

Our GWMA in 2015 did not include the Japanese or Chinese discovery panels. Furthermore, since 2015, our respective groups have undertaken genome-wide genotyping of substantially expanded Canadian, Italian, UK, and US cohorts. Therefore, we present an updated GWMA of PBC that includes these expanded cohorts, as well as the Japanese and Chinese discovery panels. In this study, we aimed to: i) capitalise on the increased sample size to discover additional risk loci for PBC; ii) explore population-specific genetic heterogeneity at known and newly identified risk loci; iii) integrate GWMA statistics with publicly available epigenetic, gene expression, and proteomic datasets to prioritise causal variants and candidate genes; and iv) use these candidate genes for in silico drug efficacy screening to identify agents potentially suitable for re-purposing to PBC.

Materials and methods

Participants and genotyping are summarised in Table 1 and detailed in the supplementary information. Written informed consent was obtained from each participant. The research conformed to the ethical guidelines of the 1975 Declaration of Helsinki.

Table 1.

Discovery panels included in the current study.

Panel (Ref) Cases Controls Variants Platform
European panels
 ‘Old’ Italian (2) 444 901 13,113,694 Illumina Human610-Quad (Cases), Illumina 1M-Duo (Controls)
 WTCCC3 (3) 1,816 5,155 12,881,032 Illumina Human-660 W Quad (Cases), Illumina 1M-Duo (Controls)
 ‘New’ Canadian-UK 4,615 9,233 8,656,760 Illumina HumanCoreExome
 ‘New’ Italian 255 579 9,264,788 Illumina HumanCoreExome
 ‘New’ US 891 621 9,964,354 Illumina Infinium Global Screening Array (GSA) v1
 European combined
8,021
16,489
5,186,747
-
Asian panels
 Japanese (4) 1,377 1,495 7,308,269 Affymetrix Axiom Genome-Wide ASI 1
 Chinese (5) 1,118 2,788 6,934,908 HumanOmniZhongHua-8
 Asian combined
2,495
4,283
5,347,815
-
All combined 10,516 20,772 2,817,608 -

WTCCC3, Wellcome Trust Case-Control Consortium 3.

Number of variants following pre- and post-imputation quality control.

Quality control

For the European and Japanese panels, quality control (QC) checks were performed at Newcastle University, UK, using the software package PLINK.7 Specific QC thresholds to determine outliers were based on visual inspection and varied by panel. For the European panels, we first removed variants with minor allele frequency (MAF) <0.01; genotype call rate <97% (<95% for the ‘old’ Italian, WTCCC3, and ‘new’ US panels); or significant deviation from Hardy Weinberg Equilibrium (HWE) (p <10−6). We then removed samples with rates of missing data >2% (>4% for the new US panel); whole-genome heterozygosity >3.25 standard deviations from the mean; apparent gender discrepancies (based on X-chromosomal heterozygosity >0.2 for men and <0.2 for women); estimated proportion of identity-by-descent sharing with another sample >0.1 (based on subsets of between 38,000 and 97,000 variants pruned for linkage disequilibrium [LD]); or that did not cluster with the CEU HapMap2 population (based on visual inspection of the first 2 principal components). For the Japanese panel, we used the dataset described in Kawashima et al. (2012),4 except for the additional removal of 4 cases and 10 controls with apparent gender discrepancies.

All samples recruited in China were processed and analysed on Chinese servers to comply with the Regulation of the People's Republic of China on the Administration of Human Genetic Resources. Thus, for the Chinese panel, QC checks were undertaken on a local server in Shanghai, China. Variants were removed with MAF <0.5%, genotype call rate <95%, or deviation from HWE in controls p ≤1x10−6. Samples were removed with rates of missing data ≥5% or pairwise identity-by-state, PI_HAT >0.25. Population outliers were identified for exclusion using principal component analysis.

Genome-wide imputation and post-imputation quality control

For the European and Japanese panels, we used the autosomal variants and samples passing QC to carry out genome-wide imputation within each individual panel using the Michigan Imputation Server with Eagle2 phasing,8 informed by the 1000 Genomes Phase 3 reference panel. Following imputation, we discarded variants with imputation R2 <0.5; non-unique alleles at the same position; or imputation call rate <90% (based on assigning genotypes according to the most likely genotype call and setting genotypes to missing if the most likely genotype call had posterior probability <0.9). We also used the resulting common set of imputed variants to check for sample duplicates/relationships across the European panels (based on estimated identity-by-descent sharing using 25,873 variants pruned for LD) and removed 1 person from each of the 137 identified relative pairs.

For the Chinese panel, genome-wide imputation was undertaken on a local server in Shanghai, China, using SHAPEIT9 and IMPUTE2,10 and the 1000 Genomes Phase 3 reference panel. Following imputation, we discarded variants with call rates <95% (having set genotypes to missing if the most likely genotype call had posterior probability <0.9), MAF <0.01, or HWE p <1×10-6 in controls. The resulting imputation summary statistics (log odds ratios [lnORs], standard errors, and p values) were submitted without individual-level data to Newcastle, UK, for meta-analysis with the other panels.

Statistical analysis of European and Japanese cohorts

Within each panel, we performed association analysis of the genome-wide imputed data using logistic regression of disease phenotype on single nucleotide polymorphism (SNP) genotype (coded 0,1,2) in PLINK,7 with the first 10 principal components (from a pruned set of SNPs with the HLA region removed) included as covariates to correct for population stratification. (The rationale for removing the HLA region was that inclusion of SNPs in this region would risk generating components that explain variation primarily caused by strong HLA-disease association, rather than population stratification.) For all but the new Canadian-UK panel, the resulting genomic control (GC) inflation factor λ was modest (<1.026); therefore, we carried out GC correction within each panel by multiplying the standard error (SE) of the estimated lnOR for each SNP by λ. For the new Canadian-UK panel, λ was somewhat inflated at 1.091; therefore, we re-analysed the new Canadian-UK data using a logistic mixed model score test (including the first 10 principal components as covariates) as implemented in the GMMAT package,11 resulting in a slightly deflated λ of 0.971. The SE of the estimated lnOR for each SNP from PLINK was then (conservatively) adjusted to match that implied by the GMMAT test statistic. Specifically, we multiplied the PLINK-derived SE for each SNP by a SNP-specific factor γ, where γ was chosen so that the resulting χ2 test statistic (lnOR/γSE)2 for that SNP had a p value equal to the p value from GMMAT. GC correction was also performed for the Chinese summary statistics (λ = 1.050) by multiplying the SE of the estimated lnOR for each SNP by λ.

Meta-analysis of European, Asian, and combined cohorts

We used the software package META12 to perform fixed-effect meta-analysis of the resulting lnORs and adjusted SEs from i) the 5 European panels; ii) the 2 Asian panels; and (3) all 7 panels, in each case restricting the analysis to variants that (following post-imputation QC) appeared within all panels. Within each meta-analysed set (European, Asian, and combined), a further GC correction was performed (to adjust for the inflation factors of 1.041, 1.033, and 1.080, seen within the European, Asian, and combined cohorts, respectively) to produce the final set of genome-wide results. Specifically, as for the individual panels above, the SE of the final lnOR for each SNP was multiplied by √λ, and the test statistic and p value were re-calculated accordingly. This use of “double” GC correction might be considered overly conservative, given that part of the observed inflation could be due to polygenicity. We explored this using LD score regression (LDSR)13 to compare our original results with those obtained using no GC (or GMMAT-derived) correction at all. We also compared our results from all panels combined with those obtained using trans-ethnic meta-regression analysis as implemented in the software package MR-MEGA14 (see supplementary information for details).

Prioritisation of candidate causal variants and candidate genes

We used the FINEMAP15 package and Conditional and Joint Analysis (COJO)16 implemented within GCTA17 to refine and look for independent associations within genome-wide significant risk loci. We used FINEMAP to construct ‘credible sets’ of variants most likely to be causal in PBC. We used the ENSEMBL Variant Effect Predictor,18 FUMA (Functional Mapping and Annotation) GWAS19 platform, and reference panels from the Avon Longitudinal Study of Parents and Children (ALSPAC, http://www.bristol.ac.uk/alspac/)20 and the INTERVAL study (http://www.donorhealth-btru.nihr.ac.uk/studies/interval-study/)21 for mapping and functional annotation of the first set of ‘credible causal variants’ at each risk locus.

Adapting the approach of Barbeira et al. (2018),22 we used the MetaXcan package; our European GWMA summary statistics; and reference panels from ALSPAC, the Genotype-Tissue Expression (GTEx) project (https://gtexportal.org/),23 and the INTERVAL study to derive genome-wide genetic prediction models of DNA methylation, gene expression, and serum protein levels in cases and controls. We used these models to correlate predicted DNA methylation, gene expression, and serum protein levels with disease status in methylome-wide, transcriptome-wide, and serum proteome-wide association studies (MWAS, TWAS, and PWAS, respectively).

We used the moloc package24 to look for co-localisation of association signals from our GWMA of the European panels with those derived from mapping of methylation, expression, and protein-quantitative trait loci (mQTLs, eQTLs, pQTLs) in ALSPAC, the GTEx project, and the INTERVAL study, respectively. Finally, we used the DEPICT package25 to prioritise the most likely causal gene at risk loci based on gene function.

Enrichment analysis

We used the STRING Database26 to look for enrichment of protein-protein interactions and functional annotations amongst candidate genes; and the DAVID resource27 to look for enrichment of KEGG pathways by genes with minimum pGWMA <0.01.

Network-based in silico drug efficacy screening

We employed the approach of Guney et al. (2016)28 in which known drug targets and candidate genes for a disease are used to estimate a drug-disease proximity measure, z, that quantifies the closeness (or proximity) of the drug and disease gene networks, respectively, correcting for the known biases of the interactome. For this analysis, we used the drug targets listed in DrugBank (https://www.drugbank.com/, accessed January 2021) and candidate genes for PBC prioritised as above. See the supplementary information for details.

Results

GWMA identifies 21 additional genome-wide significant risk loci for PBC

Following QC, the European panels consisted of 5,186,747 variants across 8,021 cases and 16,489 controls; Asian panels, 5,347,815 variants across 2,495 cases and 4,283 controls; and all panels combined, 2,817,608 variants across 10,516 cases and 20,772 controls (Table 1). Of note, there was substantial reduction in the number of variants in all panels combined compared to the European or Asian panels. This resulted from limited overlap of variants that passed post-imputation QC in the European compared to the Asian panels, explained by our use of different genotyping platforms across cohorts, and different LD patterns in Europeans compared to Asians.

GWMA of the European panels identified 46 loci at genome-wide significance (p <5×10−8); GWMA of the Asian panels, 13 loci at genome-wide significance; and GWMA of all panels combined, 41 loci at genome-wide significance (Fig. S1). Altogether, we identified 56 genome-wide significant risk loci in one or other meta-analysis (Table S1, Fig. S2). Using COJO, we identified an additional risk locus at 19p13.11 with genome-wide significance in conditional analysis of European panels (p = 4.66×10-8), having narrowly missed this threshold in the main, unconditional analysis (p = 6.55×10-8) (Fig. S2.57). Thus, a total of 57 genome-wide significant risk loci were identified in the current study. Of these, 21 were not identified in previous studies; and 2, 1q23.1 and 11q24.3, were previously identified at suggestive rather than genome-wide significance (Tables 2A&B).4,29

Table 2.

Newly identified or newly confirmed risk loci with replicated evidence of association.

Table 2A

Lead variant in the European panels
Lead variant in the Asian panels
Lead variant in the combined panels
Locus Variant:A1/A2 p value Beta Variant:A1/A2 p value Beta Variant:A1/A2 p value Beta
Gene
Chr:BP
pperm.
SE
Chr:BP
pperm.
SE
Chr:BP

SE
2p25.1 rs891058:A/G 5.39×10-7 -0.12 rs3111414:C/G 1.75×10-4 0.17 rs13416555:G/C 2.95×10-8 -0.12
ID2 2:8,442,547 0.02 2:8,443,859 0.0017 0.04 2:8,441,735 0.02
2q21.3 rs859767:G/A 1.54×10-9 -0.14 rs842349:T/G 1.76×10-9 -0.24 rs859767:G/A 8.94×10-16 -0.16
TMEM163 2:135,341,200 0.02 2:135,342,452 <0.0001 0.04 2:135,341,200 0.02
6q21 rs58926232:G/C 6.75×10-7 0.14 rs4134466:A/G 6.71×10-7 0.20 rs742108:A/G 3.16×10-8 0.13
PRDM1 6:10,6563,612 0.03 6:106,577,368 0.0001 0.04 6:106,582,920 0.02
6q27 rs3093024:A/G 2.37×10-6 0.10 rs4709148:T/C 2.18×10-10 -0.25 rs968334:T/C 3.98×10-10 0.12
CCR6 6:167,532,793 0.0001 0.02 6:167,521,676 0.04 6:167,526,096 0.02
11q24.3 rs10893872:T/C 9.07×10-6 0.10 rs11430718:G/GA 1.11×10-6 -0.19 rs10893872:T/C 9.77×10-9 0.11
ETS1 11:128,325,553 0.02 11:128,307,445 <0.0001 0.04 11:128,325,553 0.02
14q13.2 rs712315:A/T 5.70×10-7 0.15 rs199892962:AT/A 4.36×10-6 0.20 rs799469:G/A 1.73×10-9 0.15
FAM177A1
14:35,409,701

0.03
14:35,646,404
0.0020
0.04
14:35,444,425

0.03
Table 2B
Lead variant in the European panels
Lead variant in the Asian panels
Lead variant in the combined panels
Locus Variant p value Beta Variant p value Beta Variant p value Beta
Gene
Chr:BP

SE
Chr:BP
pperm.
SE
Chr:BP

SE
1q23.1 rs945635:G/C 1.59×10-8 -0.12 rs60459521:G/C 1.25×10-3 -0.46 rs11264790:T/C 2.25×10-8 -0.11
FCRL3 1:157,670,290 0.02 1:157,147,588 0.14 1:157,636,074 0.02
1q32.1 rs55734382:T/C 2.06×10-9 -0.14 rs117214467:C/T 8.55×10-3 -0.33 rs12122721:A/G 6.95×10-7 -0.11
INAVA 1:201,019,059 0.02 1:200,436,787 0.13 1:200,984,480 0.02
2p23.3 rs34655300:T/C 5.23×10-10 0.14 rs893589:A/G 9.41×10-4 0.15 rs6711622:A/G 3.89×10-8 0.11
DNMT3A 2:25,514,333 0.02 2:25,259,442 0.05 2:25,531,350 0.02
3p24.2 rs6550965:A/C 3.65×10-14 0.16 rs6807549:T/G 1.37×10-3 0.17 rs6550965:A/C 1.50×10-14 0.15
RARB 3:25,383,587 0.02 3:24,951,404 0.05 3:25,383,587 0.02
4q24 rs7663401:C/T 2.76×10-8 -0.13 rs79109654:T/C 8.56×10-5 0.37 rs2007403:T/C 6.19×10-10 0.13
TET2 4:106,128,954 0.02 4:106,170,514 0.0040 0.09 4:106,131,210 0.02
5q21.1 rs141002831:T/TCA 1.47×10-7 0.12 rs157181:A/C 3.94×10-5 0.21 rs60643069:GA/G 2.48×10-9 0.13
ST8SIA4 5:100,202,282 0.02 5:100,103,288 0.0032 0.05 5:100,238,073 0.02
5q31.3 rs10062349:G/A 7.36×10-8 -0.12 rs3761757:A/C 7.48×10-3 -0.14 rs6874308:C/T 4.67×10-8 -0.11
NDFIP1 5:141,509,597 0.02 5:141,488,219 0.05 5:141,506,911 0.02
7p21.1 rs7805218:A/G 4.12×10-8 0.13 rs77984571:C/G 7.54×10-3 -0.14 rs7786537:C/G 1.12×10-5 -0.11
ITGB8 7:20,378,801 0.02 7:20,512,650 0.05 7:20,427,776 0.02
7q34 rs370193557:GAAT/G 1.89×10-8 0.12 rs12056141:G/A 1.05×10-3 0.18 rs370193557:G/GAAT 9.37×10-10 -0.12
ZC3HAV1 7:138,729,543 0.02 7:138,797,730 0.05 7:138,729,543 0.02
8q24.21 rs4733851:A/G 2.18×10-7 0.11 rs1902780:C/T 5.51×10-4 -0.13 rs4733851:G/A 4.98×10-8 -0.11
PVT1 8:129,264,420 0.02 8:129,211,788 0.04 8:129,264,420 0.02
9q22.33 rs11390003:GA/G 2.56×10-8 -0.15 rs10283737:G/T 1.24×10-3 0.15 rs112500293:T/C 7.63×10-9 -0.15
TRIM14 9:100,741,912 0.03 9:100,780,063 0.05 9:100,763,455 0.03
10q11.23 rs7097397:A/G 2.42×10-10 -0.14 rs76129863:T/C 4.83×10-3 0.56 rs7922169:T/G 5.47×10-8 0.11
WDFY4 10:50,025,396 0.02 10:50,437,561 0.20 10:50,045,456 0.02
11p15.5 rs58523027:TAA/T 4.00×10-8 -0.12 rs3216:C/G 8.17×10-2 -0.10 rs9667500:G/A 1.74×10-4 -0.08
IRF7 11:646,986 0.02 11:214,421 0.06 11:683,761 0.02
14q32.12 rs72699866:A/G 2.89×10-11 -0.20 rs76914265:G/C 1.16×10-4 -0.30 rs4904964:C/A 2.45×10-8 -0.12
RIN3 14:93,114,787 0.03 14:93,219,854 0.0143 0.08 14:93,099,867 0.02
16q22.1 rs79577483:G/A 1.23×10-11 0.21 rs698729:G/C 1.90×10-2 -0.12 rs111644390:TC/T 1.18×10-9 0.17
DPEP3 16:68,036,939 0.03 16:68,624,205 0.05 16:68,046,323 0.03
18q22.2 rs1808094:T/C 2.79×10-9 0.13 rs76486918:T/C 2.72×10-3 -0.91 rs1808094:T/C 1.66×10-10 0.12
CD226 18:67,526,026 0.02 18:67,081,620 0.30 18:67,526,026 0.02

Results for the lead variant at newly identified or newly confirmed risk loci with p <5×10-8 in fixed-effect meta-analysis of the European, Asian, or combined panels. (A) Evidence of association was taken to be conclusive because: i) an unequivocal association signal at the same locus was observed in both the European and the Asian panels; and ii) where the lead variant at the locus was different in the European vs. the Asian panels, permutation testing confirmed the significance of the signal in the validating dataset at pperm <0.00217 (see supplementary information and Table S1). (B) Evidence of association was taken to be strong but not conclusive because unequivocal association was evident in the European but not the Asian panels, or permutation testing was not significant at pperm <0.00217. Gene: candidate gene at the risk locus (which is not necessarily the mapped gene). A1, tested allele; A2, alternative allele; BP, base pair position; Chr, chromosome; pperm, permutation p value; OR, odds ratio.

Note that 1q23.1 and 11q24.3 were previously identified at suggestive level of significance in the study by Kawashima et al. (2017).

At 6 newly identified or newly confirmed risk loci, we considered evidence of association to be conclusive because: i) an unequivocal association signal was evident in both the European and Asian panels; and ii) where the lead variant at the locus was different in the European compared to the Asian panels, permutation testing confirmed the significance of a signal in the validating dataset, located in proximity to the primary signal in the index dataset (ppermutation <0.00217, corresponding to p <0.05 Bonferroni-corrected for 23 tests; see supplementary information for details) (Table 2A, Table S1, Fig. S2).

At 17 newly identified or newly confirmed risk loci, we considered evidence of association to be strong but not conclusive because unequivocal association was evident in the European but not the Asian panels, or permutation testing was not significant at ppermutation <0.00217 (Table 2B, Table S1, Fig. S2). We note, however, that most of these loci achieved levels of significance in the Asian panels that were suggestive for validation, including 2 loci with suggestive permutation p values (4q24, ppermutation = 0.0040; and 5q21.1, ppermutation = 0.0032).

We confirmed genome-wide significant associations at 34 of 43 previously identified risk loci for PBC – but not at 9 previously identified risk loci. Seven of these 9 loci nevertheless showed a convincing association signal, albeit at p >5×10-8 (Table S2, Fig. S3). We found no evidence of association at the 15q25.1 locus (harbouring IL16) that was discovered and validated in the Chinese GWAS by Qiu et al. (2017) 5; this is explained by the absence of a signal in the Japanese and European panels. Coverage of the 19p13.2 locus was too sparse to test association.

Using FINEMAP and COJO, we found that at most risk loci, the association signal was best explained by a single variant – but at 16 loci, it was best explained by ≥2 independent variants (Table S3). Notable examples include the 2q32.2 locus harbouring STAT4, with 3 independent variants; 3q25.33 (IL12A, 3 variants); 7q32.1 (IRF5, 2 variants); and 16p13.13 (CLEC16A, 2 variants) – all consistent with previous studies showing ≥2 independent associations at each of these loci.

We compared our original results to those obtained without GC (or GMMAT-derived) correction. As expected, without correction, all loci previously identified as genome-wide significant reached slightly higher levels of significance, while a few loci that did not reach genome-wide significance in our original analysis, now (just) did so (Fig. S4 and Table S4). We also compared our original results for all panels combined with those obtained using trans-ethnic meta-regression analysis, implemented in MR-MEGA. Results from MR-MEGA were highly concordant with those from our original analysis (Fig. S5), also providing genome-wide significant confirmation of an independent association signal at 7q32.1, which exhibited significant heterogeneity in the direction of effects between the Asian and European cohorts (Table S5 and Fig. S6).

PBC shows genetic correlation with other autoimmune conditions

Recognising that most risk loci for PBC are also risk loci for other autoimmune conditions (Table S6), we used LDSR implemented via LD Hub30 to evaluate the genetic correlation between PBC (using summary statistics from our European panels) and complex traits with GWAS summary statistics in the LD Hub database. We found significant genetic correlation between PBC and other immune-mediated inflammatory disorders, including systemic lupus erythematosus (SLE, rg = 0.54, p = 2.87×10-14), rheumatoid arthritis (RA, rg = 0.26, p = 3.77×10-5), and inflammatory bowel disease (IBD, rg = 0.23, p = 6.97×10-5) (Table S7). We were unable to test genetic correlation of PBC with autoimmune thyroid disease, Sjögren syndrome, or systemic sclerosis because GWAS summary statistics for these conditions were not available in LD Hub at the time of interrogation (19.09.2019).

The genetic architecture of PBC is broadly shared across European and Asian populations

To evaluate consistency between European and Asian signals, we applied permutation testing where warranted and standard meta-analysis measures of heterogeneity to the lead variants at each of the 56 genome-wide significant risk loci identified or confirmed in the main, unconditional analyses (Table S1). We found concordance between risk loci operating in European and Asian populations, considering i) the much smaller sample size of the Asian panels; and ii) the interrogation of different variants in the European compared to the Asian panels, for reasons given above (for a detailed commentary of each risk locus, see Fig. S2). With few exceptions, we also found concordance between the lnORs seen in the combined Asian and combined European panels (Fig. S7).

To investigate overall concordance in the genetic basis of PBC between European and Asian populations, we estimated the proportion of trait variance explained (on the liability scale) in the Japanese cohort (for which individual-level genotype data were available) by sets of variants chosen according to their p values in the European GWMA (see supplementary information). Regardless of the p value threshold and the assumed trait prevalence, variants showing some level of association in the European GWMA explained more of the trait variance than an equivalent number of randomly chosen variants – in most instances, significantly more – supporting the conclusion that loci influencing the risk of PBC in Europeans, also influence its risk in Asians (Table S8).

Thus, while equivalently powered cohorts, accurately genotyped at the same set of variants, would be required to fully address the question of population-specific genetic heterogeneity, our results provide preliminary evidence that the genetic architecture of PBC is broadly shared across European and Asian populations.

Co-localisation and DEPICT enable prioritisation of candidate genes

In functional annotation, we found that credible causal variants included missense variants in 21 genes at 14 risk loci; splice variants in 8 genes at 5 risk loci; and stop variants in 2 genes at 2 risk loci (Table S9). Few of these variants were predicted to be deleterious. Credible causal variants at all genome-wide significant risk loci mapped to chromatin interacting regions (CIRs), mQTLs, eQTLs, or pQTLs (Tables S10–12); and in the MWAS, TWAS, and PWAS, we predicted differential methylation, transcription, or translation of genes at and beyond GWMA-significant loci (Tables S13–15, Fig. S8). These observations suggest that the genetic architecture of PBC confers susceptibility to disease mainly by influencing the regulation of expression of causal genes. Therefore, we sought co-localisation of GWMA with mQTL, eQTL, or pQTL association signals, aiming to pinpoint causal variants and genes across the genome. Using moloc, we identified 251 co-localisation models with posterior probability of association ≥0.80, implicating variants and genes at 60 loci (Table S16, Fig. S8C). Of these, 28 correspond to GWMA-significant risk loci, where co-localisation models implicate candidate genes such as IL12RB2 (1p31.3), FCRL3 (1q23.1), and INAVA (1q32.1). Association at the other 32 loci did not reach genome-wide significance in the GWMA; co-localisation models nevertheless implicate highly plausible candidate genes at some of these loci, such as CCL21 (9p13.3) and IL2RB (22q12.3).

We found that candidate genes implicated by co-localisation were broadly concordant with those implicated by functional annotation of credible causal variants, and by the MWAS, TWAS, and PWAS. As in previous studies, we also observed that candidate genes at disparate risk loci are evidently related in function, e.g., IL12A (3q25.33), IL12B (5q33.3), IL12RB1 (19p13.11), and IL12RB2 (1p31.3). Therefore, we used DEPICT25 to prioritise candidate genes at genome-wide significant risk loci based on gene function. In this way, we identified 82 candidate genes with a false discovery rate (FDR) <5% across 48 loci (Table S17). As expected, genes prioritised by DEPICT overlapped with those prioritised by the other approaches (Table S18).

We used the information garnered above to finalise a list of top candidate genes at genome-wide significant risk loci (Table S18). Using STRING,26 we found these genes to be highly enriched for protein-protein interactions (p <1.0×10-16), with enrichment at FDR <5% of the following KEGG pathways: T helper (TH)1 and TH2 cell differentiation, TH17 cell differentiation, and toll-like receptor (TLR), RIG-I-like receptor (RLR), TNF, NF-κB, and JAK-STAT signalling pathways, amongst others (Fig. S9). For comparison, we undertook enrichment analysis using DAVID27 of 1,388 genes with minimum pGWMA <0.01, which identified enrichment at FDR <5% of the following KEGG pathways: antigen processing and presentation, FcγR-mediated phagocytosis, NK cell-mediated cytotoxicity, and T cell receptor, B cell receptor, PI3K-AKT, FcεRI, JAK-STAT, NF-κB, and MAPK signalling pathways, amongst others (Table S19).

In silico drug efficacy screening identifies agents potentially suitable for re-purposing to PBC

In the approach of Guney et al. (2016),28 the more negative the value of z, the closer the drug and disease gene networks. A cut-off of z ≤-0.15 is taken to show that the drug is proximal to the disease and thus, might exert pharmacological effects on it. In our analysis, we identified many agents with z ≤-0.15, which are therefore predicted to exert pharmacological effects on PBC (Table 3, Table S20). Top-ranking drugs that might be predicted to ameliorate PBC included several immunomodulators, such as ustekinumab, an anti-IL-12/23 monoclonal antibody used for psoriasis and Crohn’s disease (z = -4.757); belatacept, a CTLA-4 fusion protein used in organ transplantation (z = -4.709); and abatacept, a CTLA-4 fusion protein used for RA, juvenile idiopathic arthritis (JIA), and psoriatic arthritis (z = -4.603). Of interest, other top-ranking agents include the retinoids etretinate and its metabolite acitretin, both of which are used for the treatment of psoriasis (z = -3.879 and z = -4.548, respectively). Top-ranking drugs that might be predicted to exacerbate PBC included the pharmacological interferons, such as interferon alfa-2a and interferon beta-1b (z = -2.748 and z = -2.688, respectively). Amongst recognised treatments for PBC, fenofibrate scored z = -0.986; bezafibrate, z = -0.866; and OCA, z = -0.737, respectively. Thus, these drugs might be predicted to exert pharmacological effects on PBC. Conversely, UDCA scored z = +0.171, meaning it is not predicted to treat the genetically determined component of disease in PBC.

Table 3.

In silico drug efficacy screening.

Drug name z p value Description
Ustekinumab -4.757 9.82×10-7 Anti-IL-12/23 p40 antibody
Belatacept -4.709 1.24×10-6 IgG1 Fc/CTLA-4 fusion protein
Abatacept -4.603 2.08×10-6 IgG1 Fc/CTLA-4 fusion protein
Acitretin -4.548 2.71×10-6 Oral retinoid
Denosumab -4.416 5.03×10-6 Anti-TNFSF11 antibody
Etretinate -3.879 5.24×10-5 Oral retinoid
Tofacitinib -3.340 4.19×10-4 Janus kinase inhibitor
Basiliximab -3.320 4.50×10-4 Anti-IL2Rα antibody
Gilteritinib -3.310 4.66×10-4 Tyrosine kinase inhibitor
Fostamatinib -3.305 4.75×10-4 Tyrosine kinase inhibitor
Imatinib -3.189 7.14×10-4 Tyrosine kinase inhibitor
Dexchlorpheniramine maleate -3.182 7.31×10-4 Antihistamine
Linagliptin -3.010 1.31×10-3 Dipeptidyl Peptidase-IV Inhibitor
Brigatinib -2.961 1.53×10-3 ALK and EGFR inhibitor
Interferon alfa-2a -2.748 3.00×10-3 Alpha interferon
Interferon beta-1b -2.688 3.59×10-3 Beta interferon
Metformin -1.894 0.029 Biguanide antidiabetic agent
Fenofibrate -0.986 0.162 Fibrate, PPAR-α agonist
Bezafibrate -0.866 0.193 Fibrate, PPAR-α/δ/γ agonist
Obeticholic acid -0.737 0.231 Bile acid, FXR agonist
Rifampicin -0.627 0.265 Antibiotic
Ursodeoxycholic acid +0.171 0.568 Bile acid

Results for top-ranking agents and current treatments for primary biliary cholangitis, z being a drug-disease proximity measure, defined as z = (dc-μ)/σ where dc is the average shortest path length between the drug's targets and the nearest disease gene, and μ and σ are calculated via a randomisation procedure as described in the supplementary information. Guney et al. define a drug to be proximal to a disease if its proximity follows z ≤−0.15 (p ≤0.44), and distant otherwise.

Discussion

We report the largest GWMA of PBC undertaken to date, with a sample size four times greater than that of our previous study. In this better-powered study, we identified 21 additional genome-wide significant risk loci; showed that the genetic architecture of PBC is broadly shared across European and Asian populations; prioritised candidate genes at known and newly identified genome-wide significant risk loci; and used these candidate genes to identify medications predicted to treat the genetically determined component of disease in PBC, which might therefore be suitable for re-purposing to this condition.

Candidate genes at newly identified or newly confirmed risk loci provide additional insights into the pathogenesis of PBC (Fig. 1). Thus, INAVA (1q32.1) amplifies pattern recognition receptor (PRR) signalling; DNMT3A (2p23.3), ZC3HAV1 (7q34), and TRIM14 (9q22.33) are each involved in RLR signalling; TET2 (4q24) represses transcription of IL-6; and PVT1 (8q24.21) regulates inflammation via NF-κB and MAPK pathways. Chemokine receptor 6 (CCR6, 6q27) interacts with CCL20 in the chemotaxis of dendritic cells and lymphocytes to inflamed epithelia; ST8SIA4 (5q21.1) is required for the interaction of CCR7 with CCL21 in the trafficking of immune cells to secondary lymphatic organs; and CD226 (18q22.2) participates in lymphocyte and NK cell adhesion and signalling. Fc receptor-like protein 3 (FCRL3, 1q23.1), ID2 (2p25.1), TET2 (4q24), RARB (3p24.2), NDFIP1 (5q31.3), ITGB8 (7p21.1), and CD226 (18q22.2) are each involved in the differentiation of TH1, TH17, or regulatory T cells. As expected, enrichment analysis of candidate genes reiterated the importance of PRR, TNF, and NF-κB signalling, and TH1/TH17 cell differentiation in this disease. These findings are consistent with functional data emphasising the importance of innate immune cell hypersensitivity, chemokine signalling and immune cell trafficking, and TH1/TH17 cell polarisation in PBC pathogenesis, as summarised by Gulamhusein and Hirschfield (2020)31 in their recent review.

Fig. 1.

Fig. 1

Biological processes implicated by candidate genes Inline graphic prioritised in the current study.

(A) T and B cell activation, and differentiation of T follicular helper, TH1, TH17, and TREG cells; (B) pattern recognition receptor and TNF signalling in antigen presenting cells; and (C) signalling by the IL-12 family of cytokines. TH, T helper; TREG, regulatory T. (Figure created with BioRender.com).

There is considerable current interest in the ‘Druggable Genome’, i.e., the use of genome-wide approaches to find targets for drug discovery (for example, see the Open Targets initiative at https://www.opentargets.org/). In the current study, having prioritised candidate genes, we used network-based in silico drug efficacy screening to identify agents potentially suitable for re-purposing to PBC. Given our other findings – including genetic correlation of PBC with SLE, RA, and IBD – it is expected that the top-ranking medications should include immunomodulators already approved for the treatment of RA, JIA, IBD, MS, or psoriasis.

The evidence to support re-purposing of these immunomodulators to PBC is circumstantial yet convincing – but circumspection is required. For example, in the current study, LDSR demonstrated genetic correlation with IBD; enrichment analysis showed association with ‘TH1 and TH2 cell differentiation’; and drug efficacy screening suggested that ustekinumab, an anti-IL-12/23 monoclonal antibody used for treatment of Crohn’s disease, might exert pharmacological effects on PBC. Therefore, it is notable that ustekinumab showed minimal effect on PBC in the trial by Hirschfield et al. (2016).32 Similarly, drug efficacy screening suggested that abatacept, a CTLA-4 fusion protein used for treatment of RA, might be effective for treatment of PBC – but abatacept showed no effect on PBC in the trial by Bowlus et al. (2019).33 A potential explanation for these discrepant observations, also expounded by Bowlus et al.,33 is that the evaluation of immunomodulators in PBC might require a change in clinical trial design. Thus, immunomodulators might require immunological rather than cholestatic endpoints; might be more effective in early disease, before the cholestatic liver injury predominates; and might require combined treatment of both the autoimmune and cholestatic injuries. Re-design of clinical trials in PBC might be contentious but the use of genomic data to prioritise potential agents for PBC is not, as new treatments for PBC are needed and the druggable genome provides a framework to find them.

It is notable that in drug efficacy screening, UDCA – well-established as first-line treatment for PBC – was not predicted to be therapeutic in this condition. One possibility is that UDCA serves primarily to treat a cholestatic liver injury that is critical to disease progression but orthogonal to the genetically determined, autoimmune processes that confer risk of disease. Conversely, OCA (a potent FXR agonist) and the fibrates, bezafibrate and fenofibrate (PPAR-α/δ/γ and PPAR-α agonists, respectively), are expected to have immune-modulatory as well as anti-cholestatic effects.34,35

We acknowledge 2 major limitations of the study. First, the absence of an independent validation cohort meant we were unable to confirm several newly identified risk loci. Other strategies, such as cross-phenotype meta-analysis, may be required for external validation of these loci. Second, the use of different genotyping platforms across cohorts meant that at many risk loci, the lead variant in the European panels was not represented in the Asian panels, or vice versa. This, together with marked disparity in the sample size of the European vs. the Asian panels, meant that we were unable to fully address the question of population-specific genetic heterogeneity.

In conclusion, our large, trans-ethnic GWMA of PBC has identified additional risk loci; found little evidence for population-specific genetic heterogeneity; and, through functional annotation of credible causal variants and multi-omic analysis, allowed us to prioritise candidate genes, and thereby prioritise drugs potentially suitable for re-purposing to PBC. This study emphasises the value of genomic approaches to provide biological insight and guide the development of novel therapies.

Abbreviations

ALSPAC, Avon Longitudinal Study of Parents and Children; COJO, Conditional and Joint Analysis; eQTL, expression quantitative trait locus; FDR, false discovery rate; FUMA, functional mapping and annotation; GC, genomic control; GTEx, genotype-tissue expression; GWAS, genome-wide association study; GWMA, genome-wide meta-analysis; HLA, human leukocyte antigen; HWE, Hardy-Weinburg equilibrium; IBD, inflammatory bowel disease; JIA, juvenile inflammatory arthritis; LD, linkage disequilibrium; LDSR, linkage disequilibrium score regression; MAF, minor allele frequency; mQTL, methylation quantitative trait locus; MS, multiple sclerosis; MWAS, methylome-wide association study; OCA, obeticholic acid; OR, odds ratio; PBC, primary biliary cholangitis; pQTL, protein-quantitative trait locus; PWAS, proteome-wide association study; QC, quality control; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus; SNP, single nucleotide polymorphism; TH, T helper; TWAS, transcriptome-wide association study; UDCA, ursodeoxycholic acid.

Financial support

HJC is funded by a Wellcome Trust Senior Research Fellowship in Basic Biomedical Science (102858/Z/13/Z). JJF is funded by a BBSRC DTP studentship (BB/M011186/1). The University of Cambridge has received salary support in respect of RNS from the NHS in the East of England through the Clinical Academic Reserve. KNL was supported by the NIH, DK80670. CIA is a Cancer Prevention Research Institute of Texas (CPRIT) Established Scholar and is supported by RR170048 and CA186566. AG, MC, and PI are supported by unrestricted research funding from AMAF Monza ONLUS and AIRCS, and partially supported by the Italian Ministry of University and Research (MIUR) - Department of Excellence project PREMIA (PREcision MedIcine Approach: bringing biomarker research to clinic). KAS is supported by the Sherman Family Chair in Genomic Medicine and a Foundation grant from the Canadian Institutes for Health Research (353710) and an Ontario Research Fund award (RE-09090). MN is funded by a Grant-in-Aid for Clinical Research from the National Hospital Organization and grants from Japan Society for the Promotion of Science (26293181, 17H04169). KT and MN are funded by grants from Japan Agency for Medical Research and Development (AMED) (JP20km0405205 and JP20km0405501). GFM was funded by a post-doctoral fellowship from the National Institute for Health Research (NIHR) Rare Diseases – Translational Research Collaboration (RD-TRC) and is now funded by a Clinician Academic Research Partnership (CARP) award from the Medical Research Council (MRC), UK. The MRC, UK (Grant ref: 217065/Z/19/Z) and the University of Bristol provide core support for ALSPAC, and the ALSPAC GWAS data were generated by Sample Logistics and Genotyping Facilities at Wellcome Sanger Institute and LabCorp (Laboratory Corporation of America) using support from 23andMe. A comprehensive list of grants funding is available on the ALSPAC website (http://www.bristol.ac.uk/alspac/external/documents/grant-acknowledgements.pdf). The research included in the current study involved collection of genotype data from the ARIES mothers funded by the Wellcome Trust (WT088806), and collection of the ARIES methylation data funded by the BBSRC (BBI025751/1 and BB/I025263/1). UK-PBC was funded by a Stratified Medicine award from the MRC, UK (MR/L001489/1).

Authors’ contributions

HJC, DEJ, RNS, KNL, PI, MFS, KAS, CIA, XM, MN and GFM conceived and planned the project. KNL, PI, MFS, KAS, XM, MN and GFM directed recruitment, sample collection and genotyping. HJC, JJF, KU, RD and CIA carried out the analyses. All authors contributed to interpretation of the results. HJC and GFM wrote the manuscript. All authors provided critical feedback and shaped the analysis and manuscript.

Data availability statement

Following publication, summary statistics from the current study will be deposited with the European Genome-phenome Archive.

Conflicts of interest

GMH has consulted and/or been a speaker for Intercept, Genfit, Cymabay, GSK, and Falk. RNS and GFM have each received research funding from Intercept Pharmaceuticals. HJC, JJF, KU, RD, YA, YH, MK, NN, S-SK, OG, YK, MN, KT, RT, YS, ZL, BDJ, EJA, AG, MC, RA, AC, MdA, AB, JH, MARF, DS, DEJ, SF, AS, VLM, KNL, CIA, MFS, PI, KAS, XM and MN report no conflicts of interest.

Please refer to the accompanying ICMJE disclosure forms for further details.

Acknowledgements

We are grateful to Dr Carl Anderson at the Wellcome Sanger Institute for facilitating genotyping of the UK-PBC Research Cohort. AG, MC, and PI are members of the European Reference Network on Hepatological Diseases (ERN RARE-LIVER). We are grateful to all the families who took part in ALSPAC, the midwives for their help in recruiting them, and the whole ALSPAC team, which includes interviewers, computer and laboratory technicians, clerical workers, research scientists, volunteers, managers, receptionists, and nurses. We are grateful to all the people with PBC enrolled in the respective PBC resource cohorts included in the current study.

Footnotes

Supplementary data to this article can be found online at https://doi.org/10.1016/j.jhep.2021.04.055.

Contributor Information

George F. Mells, Email: gfm26@cam.ac.uk.

Canadian PBC Consortium:

Katherine A. Siminovitch, Gideon M. Hirschfield, Andrew Mason, Catherine Vincent, Gang Xie, and Jinyi Zhang

Chinese PBC Consortium:

Ruqi Tang, Xiong Ma, Zhiqiang Li, and Yongyong Shi

Italian PBC Study Group:

Andrea Affronti, Piero L. Almasio, Domenico Alvaro, Pietro Andreone, Angelo Andriulli, Francesco Azzaroli, Pier Maria Battezzati, Antonio Benedetti, MariaConsiglia Bragazzi, Maurizia Brunetto, Savino Bruno, Vincenza Calvaruso, Vincenzo Cardinale, Giovanni Casella, Nora Cazzagon, Antonio Ciaccio, Barbara Coco, Agostino Colli, Guido Colloredo, Massimo Colombo, Silvia Colombo, Laura Cristoferi, Carmela Cursaro, Lory Saveria Crocè, Andrea Crosignani, Daphne D’Amato, Francesca Donato, Gianfranco Elia, Luca Fabris, Stefano Fagiuoli, Carlo Ferrari, Annarosa Floreani, Andrea Galli, Edoardo Giannini, Ignazio Grattagliano, Pietro Lampertico, Ana Lleo, Federica Malinverno, Clara Mancuso, Fabio Marra, Marco Marzioni, Sara Massironi, Alberto Mattalia, Luca Miele, Chiara Milani, Lorenzo Morini, Filomena Morisco, Luigi Muratori, Paolo Muratori, Grazia A. Niro, Sarah O’Donnell, Antonio Picciotto, Piero Portincasa, Cristina Rigamonti, Vincenzo Ronca, Floriano Rosina, Giancarlo Spinzi, Mario Strazzabosco, Mirko Tarocchi, Claudio Tiribelli, Pierluigi Toniutto, Luca Valenti, Maria Vinci, and Massimo Zuin

Japan-PBC-GWAS Consortium:

Hitomi Nakamura, Seigo Abiru, Shinya Nagaoka, Atsumasa Komori, Hiroshi Yatsuhashi, Hiromi Ishibashi, Masahiro Ito, Kiyoshi Migita, Hiromasa Ohira, Shinji Katsushima, Atsushi Naganuma, Kazuhiro Sugi, Tatsuji Komatsu, Tomohiko Mannami, Kouki Matsushita, Kaname Yoshizawa, Fujio Makita, Toshiki Nikami, Hideo Nishimura, Hiroshi Kouno, Hirotaka Kouno, Hajime Ota, Takuya Komura, Yoko Nakamura, Masaaki Shimada, Noboru Hirashima, Toshiki Komeda, Keisuke Ario, Makoto Nakamuta, Tsutomu Yamashita, Kiyoshi Furuta, Masahiro Kikuchi, Noriaki Naeshiro, Hironao Takahashi, Yutaka Mano, Seiji Tsunematsu, Iwao Yabuuchi, Yusuke Shimada, Kazuhiko Yamauchi, Rie Sugimoto, Hironori Sakai, Eiji Mita, Masaharu Koda, Satoru Tsuruta, Hiroshi Kamitsukasa, Takeaki Sato, Naohiko Masaki, Tatsuro Kobata, Nobuyoshi Fukushima, Yukio Ohara, Toyokichi Muro, Eiichi Takesaki, Hitoshi Takaki, Tetsuo Yamamoto, Michio Kato, Yuko Nagaoki, Shigeki Hayashi, Jinya Ishida, Yukio Watanabe, Masakazu Kobayashi, Michiaki Koga, Takeo Saoshiro, Michiyasu Yagura, Keisuke Hirata, Atsushu Tanaka, Hajime Takikawa, Mikio Zeniya, Masanori Abe, Morikazu Onji, Shuichi Kaneko, Masao Honda, Kuniaki Arai, Teruko Arinaga-Hino, Etsuko Hashimoto, Makiko Taniai, Takeji Umemura, Satoru Joshita, Kazuhiko Nakao, Tatsuki Ichikawa, Hidetaka Shibata, Satoshi Yamagiwa, Masataka Seike, Koichi Honda, Shotaro Sakisaka, Yasuaki Takeyama, Masaru Harada, Michio Senju, Osamu Yokosuka, Tatsuo Kanda, Yoshiyuki Ueno, Kentaro Kikuchi, Hirotoshi Ebinuma, Takashi Himoto, Michio Yasunami, Kazumoto Murata, Masashi Mizokami, Kazuhito Kawata, Shinji Shimoda, Yasuhiro Miyake, Akinobu Takaki, Kazuhide Yamamoto, Katsuji Hirano, Takafumi Ichida, Akio Ido, Hirohito Tsubouchi, Kazuaki Chayama, Kenichi Harada, Yasuni Nakanuma, Yoshihiko Maehara, Akinobu Taketomi, Ken Shirabe, Yuji Soejima, Akira Mori, Shintaro Yagi, Shinji Uemoto, Egawa H, Tomohiro Tanaka, Noriyo Yamashiki, Sumito Tamura, Yasuhiro Sugawara, and Norihiro Kokudo

US PBC Consortium:

Brian D. Juran, Elizabeth J. Atkinson, Angela Cheung, Mariza de Andrade, Konstantinos N. Lazaridis, Naga Chalasani, Vel Luketic, Joseph Odin, Kapil Chopra, Aris Baras, Julie Horowitz, Goncalo Abecasis, Michael Cantor, Giovanni Coppola, Aris Economides, Luca A. Lotta, John D. Overton, Jeffrey G. Reid, Alan Shuldiner, Christina Beechert, Caitlin Forsythe, Erin D. Fuller, Zhenhua Gu, Michael Lattari, Alexander Lopez, John D. Overton, Thomas D. Schleicher, Maria Sotiropoulos Padilla, Karina Toledo, Louis Widom, Sarah E. Wolf, Manasi Pradhan, Kia Manoochehri, Ricardo H. Ulloa, Xiaodong Bai, Suganthi Balasubramanian, Leland Barnard, Andrew Blumenfeld, Gisu Eom, Lukas Habegger, Alicia Hawes, Shareef Khalid, Jeffrey G. Reid, Evan K. Maxwell, William Salerno, Jeffrey C. Staples, Marcus B. Jones, and Lyndon J. Mitnaul

UK-PBC Consortium:

Richard Sturgess, Christopher Healey, Andrew Yeoman, Anton VJ. Gunasekera, Paul Kooner, Kapil Kapur, V. Sathyanarayana, Yiannis Kallis, Javaid Subhani, Rory Harvey, Roger McCorry, Paul Rooney, David Ramanaden, Richard Evans, Thiriloganathan Mathialahan, Jaber Gasem, Christopher Shorrock, Mahesh Bhalme, Paul Southern, Jeremy A. Tibble, David A. Gorard, Susan Jones, George Mells, Victoria Mulcahy, Brijesh Srivastava, Matthew R. Foxton, Carole E. Collins, David Elphick, Mazn Karmo, Francisco Porras-Perez, Michael Mendall, Tom Yapp, Minesh Patel, Roland Ede, Joanne Sayer, James Jupp, Neil Fisher, Martyn J. Carter, Konrad Koss, Jayshri Shah, Andrzej Piotrowicz, Glyn Scott, Charles Grimley, Ian R. Gooding, Simon Williams, Judith Tidbury, Guan Lim, Kuldeep Cheent, Sass Levi, Dina Mansour, Matilda Beckley, Coral Hollywood, Terry Wong, Richard Marley, John Ramage, Harriet M. Gordon, Jo Ridpath, Theodore Ngatchu, Vijay Paul Bob Grover, Ray G. Shidrawi, George Abouda, L. Corless, Mark Narain, Ian Rees, Ashley Brown, Simon Taylor-Robinson, Joy Wilkins, Leonie Grellier, Paul Banim, Debasish Das, Michael A. Heneghan, Howard Curtis, Helen C. Matthews, Faiyaz Mohammed, Mark Aldersley, Raj Srirajaskanthan, Giles Walker, Alistair McNair, Amar Sharif, Sambit Sen, George Bird, Martin I. Prince, Geeta Prasad, Paul Kitchen, Adrian Barnardo, Chirag Oza, Nurani N. Sivaramakrishnan, Prakash Gupta, Amir Shah, Chris DJ. Evans, Subrata Saha, Katharine Pollock, Peter Bramley, Ashis Mukhopadhya, Stephen T. Barclay, Natasha McDonald, Andrew J. Bathgate, Kelvin Palmer, John F. Dillon, Simon M. Rushbrook, Robert Przemioslo, Chris McDonald, Andrew Millar, Cheh Tai, Stephen Mitchell, Jane Metcalf, Syed Shaukat, Mary Ninkovic, Udi Shmueli, Andrew Davis, Asifabbas Naqvi, Tom JW. Lee, Stephen Ryder, Jane Collier, Howard Klass, Matthew E. Cramp, Nichols Sharer, Richard Aspinall, Deb Ghosh, Andrew C. Douds, Jonathan Booth, Earl Williams, Hyder Hussaini, John Christie, Steven Mann, Douglas Thorburn, Aileen Marshall, Imran Patanwala, Aftab Ala, Julia Maltby, Ray Matthew, Chris Corbett, Sam Vyas, Saket Singhal, Dermot Gleeson, Sharat Misra, Jeff Butterworth, Keith George, Tim Harding, Andrew Douglass, Harriet Mitchison, Simon Panter, Jeremy Shearman, Gary Bray, Michael Roberts, Graham Butcher, Daniel Forton, Zahid Mahmood, Matthew Cowan, Debashis Das, Chin Lye Ch’ng, Mesbah Rahman, Gregory C.A. Whatley, Emma Wesley, Aditya Mandal, Sanjiv Jain, Stephen P. Pereira, Mark Wright, Palak Trivedi, Fiona H. Gordon, Esther Unitt, Altaf Palejwala, Andrew Austin, Vishwaraj Vemala, Allister Grant, Andrew D. Higham, Alison Brind, Ray Mathew, Mark Cox, Subramaniam Ramakrishnan, Alistair King, Simon Whalley, Jocelyn Fraser, S.J. Thomson, Andrew Bell, Voi Shim Wong, Richard Kia, Ian Gee, Richard Keld, Rupert Ransford, James Gotto, and Charles Millson

Supplementary data

The following are the supplementary data to this article:

Multimedia component 1
mmc1.pdf (10.8MB, pdf)
Multimedia component 2
mmc2.zip (119.3KB, zip)
Multimedia component 3
mmc3.pdf (173.5KB, pdf)
Multimedia component 4
mmc4.pdf (2MB, pdf)
Multimedia component 5
mmc5.xlsx (1.8MB, xlsx)

References

  • 1.Hirschfield G.M., Liu X., Xu C., Lu Y., Xie G., Lu Y., et al. Primary biliary cirrhosis associated with HLA, IL12A, and IL12RB2 variants. N Engl J Med. 2009;360:2544–2555. doi: 10.1056/NEJMoa0810440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Liu X., Invernizzi P., Lu Y., Kosoy R., Lu Y., Bianchi I., et al. Genome-wide meta-analyses identify three loci associated with primary biliary cirrhosis. Nat Genet. 2010;42:658–660. doi: 10.1038/ng.627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mells G.F., Floyd J.A., Morley K.I., Cordell H.J., Franklin C.S., Shin S.Y., et al. Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis. Nat Genet. 2011;43:329–332. doi: 10.1038/ng.789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kawashima M., Hitomi Y., Aiba Y., Nishida N., Kojima K., Kawai Y., et al. Genome-wide association studies identify PRKCB as a novel genetic susceptibility locus for primary biliary cholangitis in the Japanese population. Hum Mol Genet. 2017;26:650–659. doi: 10.1093/hmg/ddw406. [DOI] [PubMed] [Google Scholar]
  • 5.Qiu F., Tang R., Zuo X., Shi X., Wei Y., Zheng X., et al. A genome-wide association study identifies six novel risk loci for primary biliary cholangitis. Nat Commun. 2017;8:14828. doi: 10.1038/ncomms14828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cordell H.J., Han Y., Mells G.F., Li Y., Hirschfield G.M., Greene C.S., et al. International genome-wide meta-analysis identifies new primary biliary cirrhosis risk loci and targetable pathogenic pathways. Nat Commun. 2015;6:8019. doi: 10.1038/ncomms9019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Purcell S., Neale B., Todd-Brown K., Thomas L., Ferreira M.A., Bender D., et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–575. doi: 10.1086/519795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Loh P.R., Danecek P., Palamara P.F., Fuchsberger C., Reshef Y.A., Finucane H.K., et al. Reference-based phasing using the haplotype reference consortium panel. Nat Genet. 2016;48:1443–1448. doi: 10.1038/ng.3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Delaneau O., Marchini J., Zagury J.F. A linear complexity phasing method for thousands of genomes. Nat Methods. 2011;9:179–181. doi: 10.1038/nmeth.1785. [DOI] [PubMed] [Google Scholar]
  • 10.Howie B.N., Donnelly P., Marchini J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. Plos Genet. 2009;5 doi: 10.1371/journal.pgen.1000529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chen H., Wang C., Conomos M.P., Stilp A.M., Li Z., Sofer T., et al. Control for population structure and relatedness for binary traits in genetic association studies via logistic mixed models. Am J Hum Genet. 2016;98:653–666. doi: 10.1016/j.ajhg.2016.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Liu J.Z., Tozzi F., Waterworth D.M., Pillai S.G., Muglia P., Middleton L., et al. Meta-analysis and imputation refines the association of 15q25 with smoking quantity. Nat Genet. 2010;42:436–440. doi: 10.1038/ng.572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bulik-Sullivan B.K., Loh P.R., Finucane H.K., Ripke S., Yang J., Schizophrenia Working Group of the Psychiatric Genomics C, et al. LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat Genet. 2015;47:291–295. doi: 10.1038/ng.3211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Magi R., Horikoshi M., Sofer T., Mahajan A., Kitajima H., Franceschini N., et al. Trans-ethnic meta-regression of genome-wide association studies accounting for ancestry increases power for discovery and improves fine-mapping resolution. Hum Mol Genet. 2017;26:3639–3650. doi: 10.1093/hmg/ddx280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Benner C., Spencer C.C., Havulinna A.S., Salomaa V., Ripatti S., Pirinen M. FINEMAP: efficient variable selection using summary data from genome-wide association studies. Bioinformatics. 2016;32:1493–1501. doi: 10.1093/bioinformatics/btw018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yang J., Ferreira T., Morris A.P., Medland S.E., Genetic Investigation of ATC, Replication DIG, et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat Genet. 2012;44:369–375. doi: 10.1038/ng.2213. S361-363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yang J., Lee S.H., Goddard M.E., Visscher P.M. GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet. 2011;88:76–82. doi: 10.1016/j.ajhg.2010.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.McLaren W., Gil L., Hunt S.E., Riat H.S., Ritchie G.R., Thormann A., et al. The ensembl variant effect predictor. Genome Biol. 2016;17:122. doi: 10.1186/s13059-016-0974-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Watanabe K., Taskesen E., van Bochoven A., Posthuma D. Functional mapping and annotation of genetic associations with FUMA. Nat Commun. 2017;8:1826. doi: 10.1038/s41467-017-01261-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Relton C.L., Gaunt T., McArdle W., Ho K., Duggirala A., Shihab H., et al. Data resource profile: accessible resource for integrated epigenomic studies (ARIES) Int J Epidemiol. 2015;44:1181–1190. doi: 10.1093/ije/dyv072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sun B.B., Maranville J.C., Peters J.E., Stacey D., Staley J.R., Blackshaw J., et al. Genomic atlas of the human plasma proteome. Nature. 2018;558:73–79. doi: 10.1038/s41586-018-0175-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Barbeira A.N., Dickinson S.P., Bonazzola R., Zheng J., Wheeler H.E., Torres J.M., et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat Commun. 2018;9:1825. doi: 10.1038/s41467-018-03621-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Consortium G.T. The genotype-tissue expression (GTEx) project. Nat Genet. 2013;45:580–585. doi: 10.1038/ng.2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Giambartolomei C., Zhenli Liu J., Zhang W., Hauberg M., Shi H., Boocock J., et al. A Bayesian framework for multiple trait colocalization from summary association statistics. Bioinformatics. 2018;34:2538–2545. doi: 10.1093/bioinformatics/bty147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pers T.H., Karjalainen J.M., Chan Y., Westra H.J., Wood A.R., Yang J., et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat Commun. 2015;6:5890. doi: 10.1038/ncomms6890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Szklarczyk D., Gable A.L., Lyon D., Junge A., Wyder S., Huerta-Cepas J., et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47:D607–D613. doi: 10.1093/nar/gky1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Huang da W., Sherman B.T., Lempicki R.A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37:1–13. doi: 10.1093/nar/gkn923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Guney E., Menche J., Vidal M., Barabasi A.L. Network-based in silico drug efficacy screening. Nat Commun. 2016;7:10331. doi: 10.1038/ncomms10331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tanaka A., Ohira H., Kikuchi K., Nezu S., Shibuya A., Bianchi I., et al. Genetic association of Fc receptor-like 3 polymorphisms with susceptibility to primary biliary cirrhosis: ethnic comparative study in Japanese and Italian patients. Tissue Antigens. 2011;77:239–243. doi: 10.1111/j.1399-0039.2010.01600.x. [DOI] [PubMed] [Google Scholar]
  • 30.Zheng J., Erzurumluoglu A.M., Elsworth B.L., Kemp J.P., Howe L., Haycock P.C., et al. LD Hub: a centralized database and web interface to perform LD score regression that maximizes the potential of summary level GWAS data for SNP heritability and genetic correlation analysis. Bioinformatics. 2017;33:272–279. doi: 10.1093/bioinformatics/btw613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Gulamhusein A.F., Hirschfield G.M. Primary biliary cholangitis: pathogenesis and therapeutic opportunities. Nat Rev Gastroenterol Hepatol. 2020;17:93–110. doi: 10.1038/s41575-019-0226-7. [DOI] [PubMed] [Google Scholar]
  • 32.Hirschfield G.M., Gershwin M.E., Strauss R., Mayo M.J., Levy C., Zou B., et al. Ustekinumab for patients with primary biliary cholangitis who have an inadequate response to ursodeoxycholic acid: a proof-of-concept study. Hepatology. 2016;64:189–199. doi: 10.1002/hep.28359. [DOI] [PubMed] [Google Scholar]
  • 33.Bowlus C.L., Yang G.X., Liu C.H., Johnson C.R., Dhaliwal S.S., Frank D., et al. Therapeutic trials of biologics in primary biliary cholangitis: an open label study of abatacept and review of the literature. J Autoimmun. 2019;101:26–34. doi: 10.1016/j.jaut.2019.04.005. [DOI] [PubMed] [Google Scholar]
  • 34.Chen M.L., Takeda K., Sundrud M.S. Emerging roles of bile acids in mucosal immunity and inflammation. Mucosal Immunol. 2019;12:851–861. doi: 10.1038/s41385-019-0162-4. [DOI] [PubMed] [Google Scholar]
  • 35.Christofides A., Konstantinidou E., Jani C., Boussiotis V.A. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism. 2021;114:154338. doi: 10.1016/j.metabol.2020.154338. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Multimedia component 1
mmc1.pdf (10.8MB, pdf)
Multimedia component 2
mmc2.zip (119.3KB, zip)
Multimedia component 3
mmc3.pdf (173.5KB, pdf)
Multimedia component 4
mmc4.pdf (2MB, pdf)
Multimedia component 5
mmc5.xlsx (1.8MB, xlsx)

Data Availability Statement

Following publication, summary statistics from the current study will be deposited with the European Genome-phenome Archive.

RESOURCES