Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

A genome-wide association study confirms PNPLA3 and identifies TM6SF2 and MBOAT7 as risk loci for alcohol-related cirrhosis

Abstract

Alcohol misuse is the leading cause of cirrhosis and the second most common indication for liver transplantation in the Western world1,2,3. We performed a genome-wide association study for alcohol-related cirrhosis in individuals of European descent (712 cases and 1,426 controls) with subsequent validation in two independent European cohorts (1,148 cases and 922 controls). We identified variants in the MBOAT7 (P = 1.03 × 10−9) and TM6SF2 (P = 7.89 × 10−10) genes as new risk loci and confirmed rs738409 in PNPLA3 as an important risk locus for alcohol-related cirrhosis (P = 1.54 × 10−48) at a genome-wide level of significance. These three loci have a role in lipid processing, suggesting that lipid turnover is important in the pathogenesis of alcohol-related cirrhosis.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Genome-wide association meta-analysis of 712 cases with alcohol-related cirrhosis and 1,466 controls.
Figure 2: Forest plots of odds ratios and 95% confidence intervals for the susceptibility loci for alcohol-related cirrhosis in comparison to alcohol misusers and population controls.
Figure 3: Fine-mapping analysis of the MBOAT7 association signals.

Similar content being viewed by others

References

  1. Kim, W.R., Brown, R.S., Terrault, N.A. & El-Serag, H. Burden of liver disease in the United States: summary of a workshop. Hepatology 36, 227–242 (2002).

    Article  Google Scholar 

  2. Rehm, J., Samokhvalov, A.V. & Shield, K.D. Global burden of alcoholic liver diseases. J. Hepatol. 59, 160–168 (2013).

    Article  Google Scholar 

  3. Burra, P. et al. Liver transplantation for alcoholic liver disease in Europe: a study from the ELTR (European Liver Transplant Registry). Am. J. Transplant. 10, 138–148 (2010).

    Article  CAS  Google Scholar 

  4. Teli, M.R., Day, C.P., Burt, A.D., Bennett, M.K. & James, O.F. Determinants of progression to cirrhosis or fibrosis in pure alcoholic fatty liver. Lancet 346, 987–990 (1995).

    Article  CAS  Google Scholar 

  5. Hrubec, Z. & Omenn, G.S. Evidence of genetic predisposition to alcoholic cirrhosis and psychosis: twin concordances for alcoholism and its biological end points by zygosity among male veterans. Alcohol. Clin. Exp. Res. 5, 207–215 (1981).

    Article  CAS  Google Scholar 

  6. Reed, T., Page, W.F., Viken, R.J. & Christian, J.C. Genetic predisposition to organ-specific endpoints of alcoholism. Alcohol. Clin. Exp. Res. 20, 1528–1533 (1996).

    Article  CAS  Google Scholar 

  7. Romeo, S. et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 40, 1461–1465 (2008).

    Article  CAS  Google Scholar 

  8. Tian, C., Stokowski, R.P., Kershenobich, D., Ballinger, D.G. & Hinds, D.A. Variant in PNPLA3 is associated with alcoholic liver disease. Nat. Genet. 42, 21–23 (2010).

    Article  CAS  Google Scholar 

  9. Stickel, F. et al. Genetic variation in the PNPLA3 gene is associated with alcoholic liver injury in Caucasians. Hepatology 53, 86–95 (2011).

    Article  CAS  Google Scholar 

  10. Pirazzi, C. et al. PNPLA3 has retinyl-palmitate lipase activity in human hepatic stellate cells. Hum. Mol. Genet. 23, 4077–4085 (2014).

    Article  CAS  Google Scholar 

  11. Smagris, E. et al. Pnpla3I148M knockin mice accumulate PNPLA3 on lipid droplets and develop hepatic steatosis. Hepatology 61, 108–118 (2015).

    Article  CAS  Google Scholar 

  12. Treutlein, J. et al. A genome-wide association study of alcohol dependence. Arch. Gen. Psychiatry 66, 773–784 (2009).

    Article  CAS  Google Scholar 

  13. 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. 5, e1000529 (2009).

    Article  Google Scholar 

  14. Innocenti, F. et al. Identification, replication, and functional fine-mapping of expression quantitative trait loci in primary human liver tissue. PLoS Genet. 7, e1002078 (2011).

    Article  CAS  Google Scholar 

  15. Chamorro, A.-J. et al. Systematic review with meta-analysis: the I148M variant of patatin-like phospholipase domain–containing 3 gene (PNPLA3) is significantly associated with alcoholic liver cirrhosis. Aliment. Pharmacol. Ther. 40, 571–581 (2014).

    Article  CAS  Google Scholar 

  16. Salameh, H. et al. PNPLA3 gene polymorphism is associated with predisposition to and severity of alcoholic liver disease. Am. J. Gastroenterol. 110, 846–856 (2015).

    Article  CAS  Google Scholar 

  17. Kozlitina, J. et al. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 46, 352–356 (2014).

    Article  CAS  Google Scholar 

  18. Patin, E. et al. Genome-wide association study identifies variants associated with progression of liver fibrosis from HCV infection. Gastroenterology 143, 1244–1252 (2012).

    Article  CAS  Google Scholar 

  19. Buch, S. et al. A genome-wide association scan identifies the hepatic cholesterol transporter ABCG8 as a susceptibility factor for human gallstone disease. Nat. Genet. 39, 995–999 (2007).

    Article  CAS  Google Scholar 

  20. Mells, G.F. et al. Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis. Nat. Genet. 43, 329–332 (2011).

    Article  CAS  Google Scholar 

  21. Melum, E. et al. Genome-wide association analysis in primary sclerosing cholangitis identifies two non-HLA susceptibility loci. Nat. Genet. 43, 17–19 (2011).

    Article  CAS  Google Scholar 

  22. Daly, A.K. et al. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat. Genet. 41, 816–819 (2009).

    Article  CAS  Google Scholar 

  23. Bierut, L.J. et al. A genome-wide association study of alcohol dependence. Proc. Natl. Acad. Sci. USA 107, 5082–5087 (2010).

    Article  CAS  Google Scholar 

  24. Han, S. et al. Integrating GWASs and human protein interaction networks identifies a gene subnetwork underlying alcohol dependence. Am. J. Hum. Genet. 93, 1027–1034 (2013).

    Article  CAS  Google Scholar 

  25. Treutlein, J. et al. Genome-wide association study of alcohol dependence. Arch. Gen. Psychiatry 66, 773–784 (2009).

    Article  CAS  Google Scholar 

  26. Edenberg, H.J. & Foroud, T. Genetics and alcoholism. Nat. Rev. Gastroenterol. Hepatol. 10, 487–494 (2013).

    Article  CAS  Google Scholar 

  27. Zintzaras, E., Stefanidis, I., Santos, M. & Vidal, F. Do alcohol-metabolizing enzyme gene polymorphisms increase the risk of alcoholism and alcoholic liver disease? Hepatology 43, 352–361 (2006).

    Article  CAS  Google Scholar 

  28. He, S. et al. A sequence variation (I148M) in PNPLA3 associated with nonalcoholic fatty liver disease disrupts triglyceride hydrolysis. J. Biol. Chem. 285, 6706–6715 (2010).

    Article  CAS  Google Scholar 

  29. Gijón, M.A., Riekhof, W.R., Zarini, S., Murphy, R.C. & Voelker, D.R. Lysophospholipid acyltransferases and arachidonate recycling in human neutrophils. J. Biol. Chem. 283, 30235–30245 (2008).

    Article  Google Scholar 

  30. Yamashita, A. et al. Reverse reaction of lysophosphatidylinositol acyltransferase. Functional reconstitution of coenzyme A–dependent transacylation system. J. Biol. Chem. 278, 30382–30393 (2003).

    Article  CAS  Google Scholar 

  31. Moreno-Navarrete, J.M. et al. The l-α-lysophosphatidylinositol/GPR55 system and its potential role in human obesity. Diabetes 61, 281–291 (2012).

    Article  CAS  Google Scholar 

  32. Tam, J. et al. Endocannabinoids in liver disease. Hepatology 53, 346–355 (2011).

    Article  CAS  Google Scholar 

  33. Patsenker, E. et al. Cannabinoid receptor type I modulates alcohol-induced liver fibrosis. Mol. Med. 17, 1285–1294 (2011).

    Article  CAS  Google Scholar 

  34. Liu, Y.-L. et al. TM6SF2 rs58542926 influences hepatic fibrosis progression in patients with non-alcoholic fatty liver disease. Nat. Commun. 5, 4309 (2014).

    Article  CAS  Google Scholar 

  35. Dongiovanni, P. et al. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology 61, 506–514 (2015).

    Article  CAS  Google Scholar 

  36. Speliotes, E.K. et al. Genome-wide association analysis identifies variants associated with nonalcoholic fatty liver disease that have distinct effects on metabolic traits. PLoS Genet. 7, e1001324 (2011).

    Article  CAS  Google Scholar 

  37. Nischalke, H.D. et al. A common polymorphism in the NCAN gene is associated with hepatocellular carcinoma in alcoholic liver disease. J. Hepatol. 61, 1073–1079 (2014).

    Article  CAS  Google Scholar 

  38. Di Martino, V., Sheppard, F. & Vanlemmens, C. Early liver transplantation for severe alcoholic hepatitis. N. Engl. J. Med. 366, 478–479 (2012).

    CAS  PubMed  Google Scholar 

  39. Vanlemmens, C. et al. Immediate listing for liver transplantation versus standard care for Child-Pugh stage B alcoholic cirrhosis: a randomized trial. Ann. Intern. Med. 150, 153–161 (2009).

    Article  Google Scholar 

  40. Pruim, R.J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–2337 (2010).

    Article  CAS  Google Scholar 

  41. Higgins, J.P.T. & Thompson, S.G. Quantifying heterogeneity in a meta-analysis. Stat. Med. 21, 1539–1558 (2002).

    Article  Google Scholar 

  42. Way, M. et al. Genetic variants in or near ADH1B and ADH1C affect susceptibility to alcohol dependence in a British and Irish population. Addict Biol. 20, 594–604 (2015).

    Article  CAS  Google Scholar 

  43. Trépo, E. et al. Marked 25-hydroxyvitamin D deficiency is associated with poor prognosis in patients with alcoholic liver disease. J. Hepatol. 59, 344–350 (2013).

    Article  Google Scholar 

  44. Ishak, K.G., Zimmerman, H.J. & Ray, M.B. Alcoholic liver disease: pathologic, pathogenetic and clinical aspects. Alcohol. Clin. Exp. Res. 15, 45–66 (1991).

    Article  CAS  Google Scholar 

  45. Nguyen-Khac, E. et al. Assessment of asymptomatic liver fibrosis in alcoholic patients using fibroscan: prospective comparison with seven non-invasive laboratory tests. Aliment. Pharmacol. Ther. 28, 1188–1198 (2008).

    Article  CAS  Google Scholar 

  46. Browning, B.L. & Yu, Z. Simultaneous genotype calling and haplotype phasing improves genotype accuracy and reduces false-positive associations for genome-wide association studies. Am. J. Hum. Genet. 85, 847–861 (2009).

    Article  CAS  Google Scholar 

  47. Price, A.L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    Article  CAS  Google Scholar 

  48. Roshyara, N.R. & Scholz, M. fcGENE: a versatile tool for processing and transforming SNP datasets. PLoS ONE 9, e97589 (2014).

    Article  Google Scholar 

  49. Sinnott, J.A. & Kraft, P. Artifact due to differential error when cases and controls are imputed from different platforms. Hum. Genet. 131, 111–119 (2012).

    Article  Google Scholar 

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

Download references

Acknowledgements

This study was supported by the German Ministry of Education and Research through the Virtual Liver Network (to J.H.), the PopGen 2.0 network biobank (grant 01EY1103) and institutional funds from the medical faculties of TU Dresden and Christian Albrechts University Kiel and by Swiss National Funds (grant 310030_138747 to F.S.). The Community Medicine Research network of the University of Greifswald, Germany, is funded by the Federal Ministry of Education and Research, the Ministry of Cultural Affairs and the Social Ministry of the Federal State of Mecklenburg–West Pomerania. M.M.L. and J.M. were supported by the Federal Ministry of Education and Research (BMBF GANI-MED 03152061A and BMBF 0314107), the European Union (EU-FP-7: EPC-TM and EU-FP-7-REGPOT-2010-1) and the EFRE–State Ministry of Economics (V-630-S-150-2012/132/133). S.C., M.M.N. and M. Rietschel were supported by the German Federal Ministry of Education and Research (BMBF) through the Integrated Networks IntegraMent and Sysmed Alcohol under the auspices of the e:Med Programme (grant 01ZX1314A to M.M.N. and S.C. and grant 01ZX1311A to M.M.N. and M. Rietschel). M.M.N. is a member of the DFG (Deutsche Forschungsgemeinschaft)-funded Excellence Cluster ImmunoSensation. Research by H.D.N. related to this project was funded by the Deutsche Krebshilfe (107865). The work of A.F. and D.E. was supported by the German Federal Ministry of Education and Research (BMBF) within the framework of the e:Med research and funding concept (SysInflame grant 01ZX1306A). This project received infrastructure support from DFG Excellence Cluster 306, 'Inflammation at Interfaces'. A.F. receives an endowment professorship from the Foundation for Experimental Medicine (Zurich, Switzerland). The UK research effort was funded by a PhD studentship award jointly funded by University College London and an anonymous donor. We thank colleagues from the following centers for obtaining samples from alcohol-dependent cases for genotyping: the Bexley Substance Misuse Service, South London and Maudsley National Health Service (NHS) Trust; the East Hertfordshire Community Drug Action Team; the Max Glatt Unit, Southall; Renfrew and Inverclyde Alcohol Services, Strathclyde; the Newcastle North Tyneside Drug and Alcohol Service, Tyne and Wear; and the Acute Admissions Unit and the Centre for Hepatology at the Royal Free Hospital, London. We also thank colleagues associated with the National Institute for Health Research (NIHR) Mental Health Research Network for their assistance in identifying cases, obtaining consent and collecting samples at the following NHS trusts: Sandwell Mental Health and Social Care; Northamptonshire Healthcare; Avon and Wiltshire Mental Health Partnership, Sheffield Health and Social Care; Tees Esk and Wear Valleys; Lincolnshire Partnership; Nottinghamshire Healthcare; Central and North West London; South Staffordshire and Shropshire Healthcare; Coventry and Warwickshire; and Dudley and Walsall Mental Health Partnership. We are grateful to J. Saini, K. Ruparelia, S. Montagnese, R. Kandaswamy, A. Jarram, G. Quadri and N. O'Brien for assisting with the collection and processing of samples and DNA extraction.

The Belgian research effort was supported by the Belgian Medical Genomics Initiative (BeMGI) funded by the phase VII Interuniversity Attraction Poles (IAP) program of the Belgian Federal Science Policy Office (BELSPO) and the Fund for Scientific Research–FNRS (F.R.S.-FNRS). E.T. is a Postdoctoral Researcher of the F.R.S.-FNRS, and D.F. is a Research Director of the F.R.S.-FNRS. We are grateful to O. Lemoine, D. Degré, A. Lemmers and M. Amrani for identifying cases and controls, obtaining consent and collecting samples at CUB Hôpital Erasme, Université Libre de Bruxelles. We also thank E. Quertinmont at the Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, for his help with the collection and processing of samples and DNA extraction.

We are also grateful to the Center for Information Services and High-Performance Computing (ZIH) at TU Dresden where the computations were performed on a PC cluster.

Author information

Authors and Affiliations

Authors

Contributions

S. Buch performed genotyping, meta-analysis and in silico analysis, and drafted and revised the manuscript. F.S. conceptualized the study, recruited subjects, and wrote and revised the manuscript. E.T. recruited subjects, validated the study, provided replication data, and wrote and revised the manuscript. M.W. recruited subjects, performed genotyping for the validation study and revised the manuscript. A.H. performed bioinformatics work. H.D.N. recruited and phenotyped subjects. M.B. performed expression analysis. J.R. and T.B. recruited subjects. M. Ridinger, M. Rietschel, A.M., J.F. and F.K. recruited subjects and performed phenotyping and recruitment of alcoholic controls. S.S. provided technical support and critically revised the manuscript. W.L. helped with population genetic statistics. M.S. recruited subjects and phenotyped alcoholic controls. N.S., E.A., C.D., R.S., S. Brückner, S. Zeissig and A.-M.S. recruited subjects. N.W. recruited subjects and performed phenotyping of alcoholic controls. J.D., N.C., C. Sarrazin, F.L., T.G. and P.D. recruited and phenotyped subjects. H.V. recruited the population cohort. M.M.L., J.M., F.E. and C. Schafmayer recruited and phenotyped subjects. S.C. and M.M.N. performed phenotyping and recruitment of alcoholic controls. M.N. supervised and reviewed statistical analysis. D.E. assisted with bioinformatics analysis. K.H. performed expression analysis. A.F. gave conceptual advice and bioinformatics support. S. Zopf, C.H. and C.M. recruited subjects. D.F. and M.Y.M. recruited subjects and drafted and critically revised the manuscript. J.H. conceptualized the study and analytical design, and drafted and revised the manuscript. All authors critically revised and contributed to the final manuscript.

Corresponding author

Correspondence to Felix Stickel.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Quantile-quantile plot of the primary meta-analysis of the German and UK data sets.

Samples are as outlined in Table 1. After GC adjustment, a λ value of 1.005 was obtained for the combined analysis.

Supplementary Figure 2 Manhattan plot of the adjusted meta-analysis of the German and UK data sets.

Samples are as outlined in Supplementary Table 2.

Supplementary Figure 3 Quantile-quantile plot of the adjusted meta-analysis of the German and UK data sets.

After GC adjustment, a λ value of 0.985 was obtained for the combined analysis.

Supplementary Figure 4 Fine-mapping analysis of the TM6SF2 association signals.

The –log10 (P values) are plotted against SNP genomic position based on NCBI Build 37. The known coding variant of likely functional significance, rs58542926 is highlighted in purple. The squares denote genotyped SNPs; the circles denote imputed SNPs (using 1000 Genomes Project–based imputation). SNPs are colored to reflect correlation with the most significant SNP, with red denoting the highest LD (r2 >0.8) to the lead SNP. Estimated recombination rates from 1000 Genomes Project (hg19/genomes March 2012 EUR) are plotted in blue to reflect the local LD structure. Gene annotations were obtained from the UCSC Genome Browser. The plot was generated using LocusZoom.

Supplementary Figure 5 Tissue expression of MBOAT7 and TMC4 mRNA.

Expression was tested by PCR in the human cDNA tissue panel from Takara Clontech (636742 and 636743) using transcript-specific primer pairs (MBOAT7: 5′-TCCTTGTGTCTTTCGCTCC-3′ and 5′-TACACACGGTGACCTGTCA-3′; TMC4: 5′-TGAGACCACCCAGAATTTCC-3′ and 5′-CTAGGCTTACAATGGGCCTG-3′). MBOAT7 shows a ubiquitous expression pattern in the tissues tested, albeit with lower expression in skeletal muscle. Expression of TMC4 is more selective, with no expression detected in brain or skeletal muscle.

Supplementary Figure 6 Genotype-specific relative transcript abundance in liver tissue from patients with alcohol-related cirrhosis.

No significant change in transcript abundance was seen for TMC4 across genotypes, whereas a significant increase in transcript abundance was detected for MBOAT7 in the homozygous mutant genotype (Mann-Whitney U test P = 0.0087).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 2, 3 and 5–7. (PDF 887 kb)

Supplementary Table 1: Results of the primary GWAS meta-analysis (Germany/UK).

Results of the GWAS meta-analysis of 712 cases with alcohol-related cirrhosis and 1,466 controls. Variants that entered replication genotyping are marked "SNP1" to "SNP10". SNPs are ranked by combined P value, and data are provided for the top variants through SNP10. (XLSX 534 kb)

Supplementary Table 4: Results of the secondary GWAS meta-analysis (Germany/UK) adjusted for sex, age, BMI and type 2 diabetes status.

Results of the GWAS meta-analysis adjusted for age, sex, BMI and type 2 diabetes status. Variants that entered replication genotyping are marked "SNP1" to "SNP10". SNPs are ranked by combined P value, and data are provided for the top variants through SNP10. Adjusted meta-analysis results for the replicating variants of the primary analysis are as follows: TM6SF2 (rs10401969), Pmeta = 0.000667, ORmeta = 1.87 (1.30–2.69); MBOAT7 (rs626283), Pmeta = 0.0173, ORmeta = 1.28 (1.04–1.56). These rank below SNP10 in this analysis. (XLSX 153 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Buch, S., Stickel, F., Trépo, E. et al. A genome-wide association study confirms PNPLA3 and identifies TM6SF2 and MBOAT7 as risk loci for alcohol-related cirrhosis. Nat Genet 47, 1443–1448 (2015). https://doi.org/10.1038/ng.3417

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3417

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing