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:

Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma

Abstract

Concentrations of liver enzymes in plasma are widely used as indicators of liver disease. We carried out a genome-wide association study in 61,089 individuals, identifying 42 loci associated with concentrations of liver enzymes in plasma, of which 32 are new associations (P = 10−8 to P = 10−190). We used functional genomic approaches including metabonomic profiling and gene expression analyses to identify probable candidate genes at these regions. We identified 69 candidate genes, including genes involved in biliary transport (ATP8B1 and ABCB11), glucose, carbohydrate and lipid metabolism (FADS1, FADS2, GCKR, JMJD1C, HNF1A, MLXIPL, PNPLA3, PPP1R3B, SLC2A2 and TRIB1), glycoprotein biosynthesis and cell surface glycobiology (ABO, ASGR1, FUT2, GPLD1 and ST3GAL4), inflammation and immunity (CD276, CDH6, GCKR, HNF1A, HPR, ITGA1, RORA and STAT4) and glutathione metabolism (GSTT1, GSTT2 and GGT), as well as several genes of uncertain or unknown function (including ABHD12, EFHD1, EFNA1, EPHA2, MICAL3 and ZNF827). Our results provide new insight into genetic mechanisms and pathways influencing markers of liver function.

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
Figure 2: Manhattan plots of association of SNPs with ALT, ALP and GGT in the GWAS.
Figure 3: Association of FADS1, FADS2, GCKR, HNF1A, TRIB1 and PPP1R3B loci with NMR metabonome.

Similar content being viewed by others

References

  1. Pratt, D.S. & Kaplan, M.M. Evaluation of abnormal liver-enzyme results in asymptomatic patients. N. Engl. J. Med. 342, 1266–1271 (2000).

    Article  CAS  Google Scholar 

  2. Söderberg, C. et al. Decreased survival of subjects with elevated liver function tests during a 28-year follow-up. Hepatology 51, 595–602 (2010).

    Article  Google Scholar 

  3. Xu, K. et al. Diagnostic value of serum γ-glutamyl transferase isoenzyme for hepatocellular carcinoma: a 10-year study. Am. J. Gastroenterol. 87, 991–995 (1992).

    CAS  PubMed  Google Scholar 

  4. Sattar, N. et al. Elevated alanine aminotransferase predicts new-onset type 2 diabetes independently of classical risk factors, metabolic syndrome, and C-reactive protein in the west of Scotland coronary prevention study. Diabetes 53, 2855–2860 (2004).

    Article  CAS  Google Scholar 

  5. Ioannou, G.N., Weiss, N.S., Boyko, E.J., Mozaffarian, D. & Lee, S.P. Elevated serum alanine aminotransferase activity and calculated risk of coronary heart disease in the United States. Hepatology 43, 1145–1151 (2006).

    Article  CAS  Google Scholar 

  6. Watkins, P.B. Idiosyncratic liver injury: challenges and approaches. Toxicol. Pathol. 33, 1–5 (2005).

    Article  CAS  Google Scholar 

  7. Rahmioglu, N. et al. Epidemiology and genetic epidemiology of the liver function test proteins. PLoS ONE 4, e4435 (2009).

    Article  Google Scholar 

  8. Nugent, C. & Younossi, Z.M. Evaluation and management of obesity-related nonalcoholic fatty liver disease. Nat. Clin. Pract. Gastroenterol. Hepatol. 4, 432–441 (2007).

    Article  Google Scholar 

  9. Pe'er, I., Yelensky, R., Altshuler, D. & Daly, M.J. Estimation of the multiple testing burden for genomewide association studies of nearly all common variants. Genet. Epidemiol. 32, 381–385 (2008).

    Article  Google Scholar 

  10. Yuan, X. et al. Population-based genome-wide association studies reveal six loci influencing plasma levels of liver enzymes. Am. J. Hum. Genet. 83, 520–528 (2008).

    Article  CAS  Google Scholar 

  11. Kamatani, Y. et al. Genome-wide association study of hematological and biochemical traits in a Japanese population. Nat. Genet. 42, 210–215 (2010).

    CAS  Google Scholar 

  12. The International HapMap Consortium. A second generation human haplotype map of over 3.1 million SNPs. Nature 449, 851–861 (2007).

  13. Paulusma, C.C. et al. Atp8b1 deficiency in mice reduces resistance of the canalicular membrane to hydrophobic bile salts and impairs bile salt transport. Hepatology 44, 195–204 (2006).

    Article  CAS  Google Scholar 

  14. Iwamori, M. & Domino, S.E. Tissue-specific loss of fucosylated glycolipids in mice with targeted deletion of alpha(1,2)fucosyltransferase genes. Biochem. J. 380, 75–81 (2004).

    Article  CAS  Google Scholar 

  15. LeBoeuf, R.C. et al. Mouse glycosylphosphatidylinositol-specific phospholipase D (Gpld1) characterization. Mamm. Genome 9, 710–714 (1998).

    Article  CAS  Google Scholar 

  16. Dupuis, J. et al. New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat. Genet. 42, 105–116 (2010).

    Article  CAS  Google Scholar 

  17. Voight, B.F. et al. Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat. Genet. 42, 579–589 (2010).

    Article  CAS  Google Scholar 

  18. Zeggini, E. et al. Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat. Genet. 40, 638–645 (2008).

    Article  CAS  Google Scholar 

  19. Elliott, P. et al. Genetic Loci associated with C-reactive protein levels and risk of coronary heart disease. J. Am. Med. Assoc. 302, 37–48 (2009).

    Article  CAS  Google Scholar 

  20. Bull, L.N. et al. A gene encoding a P-type ATPase mutated in two forms of hereditary cholestasis. Nat. Genet. 18, 219–224 (1998).

    Article  CAS  Google Scholar 

  21. Shin, D.M., Zhao, X.S., Zeng, W., Mozhayeva, M. & Muallem, S. The mammalian Sec6/8 complex interacts with Ca(2+) signaling complexes and regulates their activity. J. Cell Biol. 150, 1101–1112 (2000).

    Article  CAS  Google Scholar 

  22. The 1000 Genomes Projects Consortium. et al. A map of human genome variation from population-scale sequencing. Nature 467, 1061–1073 (2010).

  23. Dixon, A.L. et al. A genome-wide association study of global gene expression. Nat. Genet. 39, 1202–1207 (2007).

    Article  CAS  Google Scholar 

  24. Schadt, E.E. et al. Mapping the genetic architecture of gene expression in human liver. PLoS Biol. 6, e107 (2008).

    Article  Google Scholar 

  25. Emilsson, V. et al. Genetics of gene expression and its effect on disease. Nature 452, 423–428 (2008).

    Article  CAS  Google Scholar 

  26. Raychaudhuri, S. et al. Identifying relationships among genomic disease regions: predicting genes at pathogenic SNP associations and rare deletions. PLoS Genet. 5, e1000534 (2009).

    Article  Google Scholar 

  27. Noé, J., Stieger, B. & Meier, P.J. Functional expression of the canalicular bile salt export pump of human liver. Gastroenterology 123, 1659–1666 (2002).

    Article  Google Scholar 

  28. van Mil, S.W. et al. Benign recurrent intrahepatic cholestasis type 2 is caused by mutations in ABCB11. Gastroenterology 127, 379–384 (2004).

    Article  CAS  Google Scholar 

  29. Knisely, A.S. et al. Hepatocellular carcinoma in ten children under five years of age with bile salt export pump deficiency. Hepatology 44, 478–486 (2006).

    Article  CAS  Google Scholar 

  30. Hindorff, L.A. et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc. Natl. Acad. Sci. USA 106, 9362–9367 (2009).

    Article  CAS  Google Scholar 

  31. Chambers, J.C. et al. Genetic loci influencing kidney function and chronic kidney disease. Nat. Genet. 42, 373–375 (2010).

    Article  CAS  Google Scholar 

  32. Sabatti, C. et al. Genome-wide association analysis of metabolic traits in a birth cohort from a founder population. Nat. Genet. 41, 35–46 (2009).

    Article  CAS  Google Scholar 

  33. Gieger, C. et al. Genetics meets metabolomics: a genome-wide association study of metabolite profiles in human serum. PLoS Genet. 4, e1000282 (2008).

    Article  Google Scholar 

  34. Illig, T. et al. A genome-wide perspective of genetic variation in human metabolism. Nat. Genet. 42, 137–141 (2010).

    Article  CAS  Google Scholar 

  35. Stolk, R.P. et al. Universal risk factors for multifactorial diseases: LifeLines: a three-generation population-based study. Eur. J. Epidemiol. 23, 67–74 (2008).

    Article  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. Klomp, L.W. et al. Characterization of mutations in ATP8B1 associated with hereditary cholestasis. Hepatology 40, 27–38 (2004).

    Article  CAS  Google Scholar 

  38. Petit, J.M. et al. Specifically PNPLA3-mediated accumulation of liver fat in obese patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 95, E430–E436 (2010).

    Article  CAS  Google Scholar 

  39. Dunn, J.S. et al. Examination of PPP1R3B as a candidate gene for the type 2 diabetes and MODY loci on chromosome 8p23. Ann. Hum. Genet. 70, 587–593 (2006).

    Article  CAS  Google Scholar 

  40. 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 

  41. Saxena, R. et al. Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316, 1331–1336 (2007).

    Article  CAS  Google Scholar 

  42. Bolt, H.M. & Thier, R. Relevance of the deletion polymorphisms of the glutathione S-transferases GSTT1 and GSTM1 in pharmacology and toxicology. Curr. Drug Metab. 7, 613–628 (2006).

    Article  CAS  Google Scholar 

  43. Zhang, H., Forman, H.J. & Choi, J. γ-glutamyl transpeptidase in glutathione biosynthesis. Methods Enzymol. 401, 468–483 (2005).

    Article  CAS  Google Scholar 

  44. Wolpin, B.M. et al. Variant ABO blood group alleles, secretor status, and risk of pancreatic cancer: results from the pancreatic cancer cohort consortium. Cancer Epidemiol. Biomarkers Prev. 19, 3140–3149 (2010).

    Article  CAS  Google Scholar 

  45. Edgren, G. et al. Risk of gastric cancer and peptic ulcers in relation to ABO blood type: a cohort study. Am. J. Epidemiol. 172, 1280–1285 (2010).

    Article  Google Scholar 

  46. Lindesmith, L. et al. Human susceptibility and resistance to Norwalk virus infection. Nat. Med. 9, 548–553 (2003).

    Article  CAS  Google Scholar 

  47. Hazra, A. et al. Common variants of FUT2 are associated with plasma vitamin B12 levels. Nat. Genet. 40, 1160–1162 (2008).

    Article  CAS  Google Scholar 

  48. Heath, A.C. et al. Genetic and environmental contributions to alcohol dependence risk in a national twin sample: consistency of findings in women and men. Psychol. Med. 27, 1381–1396 (1997).

    Article  CAS  Google Scholar 

  49. Wallace, C. et al. Genome-wide association study identifies genes for biomarkers of cardiovascular disease: serum urate and dyslipidemia. Am. J. Hum. Genet. 82, 139–149 (2008).

    Article  CAS  Google Scholar 

  50. Firmann, M. et al. The CoLaus study: a population-based study to investigate the epidemiology and genetic determinants of cardiovascular risk factors and metabolic syndrome. BMC Cardiovasc. Disord. 8, 6 (2008).

    Article  Google Scholar 

  51. Kong, A. et al. Parental origin of sequence variants associated with complex diseases. Nature 462, 868–874 (2009).

    Article  CAS  Google Scholar 

  52. Watkinson, C., van Sluijs, E.M., Sutton, S., Marteau, T. & Griffin, S.J. Randomised controlled trial of the effects of physical activity feedback on awareness and behaviour in UK adults: the FAB study protocol. BMC Public Health [ISRCTN92551397] 10, 144 (2010).

    Article  Google Scholar 

  53. Kaprio, J. Twin studies in Finland 2006. Twin Res. Hum. Genet. 9, 772–777 (2006).

    Article  Google Scholar 

  54. Levy, D. et al. Genome-wide association study of blood pressure and hypertension. Nat. Genet. 41, 677–687 (2009).

    Article  CAS  Google Scholar 

  55. Löwel, H. et al. The MONICA Augsburg surveys–basis for prospective cohort studies. Gesundheitswesen 67 (suppl 1), S13–S18 (2005).

    Article  Google Scholar 

  56. Lamers, F et al. Comorbidity patterns of anxiety and depressive disorders in a large cohort study: the Netherlands Study of Depression and Anxiety (NESDA). J. Clin. Psychiatry 72, 341–348 (2011).

    Article  Google Scholar 

  57. Boomsma, D.I. et al. Netherlands Twin Register: from twins to twin families. Twin Res. Hum. Genet. 9, 849–857 (2006).

    Article  Google Scholar 

  58. Clarke, R. et al. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N. Engl. J. Med. 361, 2518–2528 (2009).

    Article  CAS  Google Scholar 

  59. Hofman, A. et al. The Rotterdam Study: 2010 objectives and design update. Eur. J. Epidemiol. 24, 553–572 (2009).

    Article  Google Scholar 

  60. Scuteri, A. et al. Genome-wide association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genet. 3, e115 (2007).

    Article  Google Scholar 

  61. Haring, R. et al. Prediction of metabolic syndrome by low serum testosterone levels in men: results from the study of health in Pomerania. Diabetes 58, 2027–2031 (2009).

    Article  CAS  Google Scholar 

  62. Spector, T.D. & MacGregor, A.J. The St. Thomas' UK Adult Twin Registry. Twin Res. 5, 440–443 (2002).

    Article  Google Scholar 

  63. 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 

  64. Bennett-Lovsey, R.M., Herbert, A.D., Sternberg, M.J. & Kelley, L.A. Exploring the extremes of sequence/structure space with ensemble fold recognition in the program Phyre. Proteins 70, 611–625 (2008).

    Article  CAS  Google Scholar 

  65. Raychaudhuri, S. et al. Identifying relationships among genomic disease regions: predicting genes at pathogenic SNP associations and rare deletions. PLoS Genet. 5, e1000534 (2009).

    Article  Google Scholar 

  66. Jimenez-Marin, A., Collado-Romero, M., Ramirez-Boo, M., Arce, C. & Garrido, J.J. Biological pathway analysis by ArrayUnlock and Ingenuity Pathway Analysis. BMC Proc. 3 (suppl. 4), S6 (2009).

    Article  Google Scholar 

  67. Schumann, G. et al. Genome-wide association and genetic functional studies identify autism susceptibility candidate 2 gene (AUTS2) in the regulation of alcohol consumption. Proc. Natl. Acad. Sci. USA 108, 7119–7124 (2011).

    Article  CAS  Google Scholar 

  68. Newton-Cheh, C. et al. Genome-wide association study identifies eight loci associated with blood pressure. Nat. Genet. 41, 666–676 (2009).

    Article  CAS  Google Scholar 

  69. Speliotes, E.K. et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 42, 937–948 (2010).

    Article  CAS  Google Scholar 

  70. Teslovich, T.M. et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466, 707–713 (2010).

    Article  CAS  Google Scholar 

  71. Inouye, M. et al. Metabonomic, transcriptomic, and genomic variation of a population cohort. Mol. Syst. Biol. 6, 441 (2010).

    Article  Google Scholar 

  72. Soininen, P. et al. High-throughput serum NMR metabonomics for cost-effective holistic studies on systemic metabolism. Analyst 134, 1781–1785 (2009).

    Article  CAS  Google Scholar 

  73. Würtz, P. et al. Characterization of systemic metabolic phenotypes associated with subclinical atherosclerosis. Mol. Biosyst. 7, 385–393 (2011).

    Article  Google Scholar 

Download references

Acknowledgements

We thank the many colleagues who contributed to collection and phenotypic characterization of the clinical samples, as well as genotyping and analysis of data. We also thank the research participants who took part in these studies. Major support for the work came from European Commission (FP5, FP6 and FP7); European Science Foundation; European Science Council; US NIH; US National Institute of Mental Health; US NIDDK; Genetic Association Information Network; US National Institute on Aging; US National Human Genome Research Institute; US NHLBI; UK NIHR; NIHR Comprehensive Biomedical Research Centre Imperial College Healthcare NHS Trust; NIHR Comprehensive Biomedical Research Centre Guy's and St. Thomas' NHS Trust; UK Biotechnology and Biological Sciences Research Council; UK MRC; British Heart Foundation; Wellcome Trust; Swiss National Science Foundation; Academy of Finland; Finnish Cardiovascular Research Foundation; Swedish Research Council; Swedish Heart-Lung Foundation; Helmholtz Zentrum München; German Research Center for Environmental Health; German Federal Ministry of Education and Research; German National Genome Research Network; Netherlands Organization for Scientific Research; Dutch Ministries of Economic Affairs, of Education, Culture and Science, for Health, Welfare and Sports; Netherlands Organization for Health Research and Development; Economic Structure Enhancing Fund of the Dutch government; Dutch Kidney Foundation; Dutch Diabetes Research Foundation; Dutch Brain Foundation; Dutch Research Institute for Diseases in the Elderly; Netherlands Genomics Initiative; Canadian Institutes for Health Research; Ontario Research Fund; The Barts and the London Charity; University Medical Center Groningen; University of Groningen; University of Oulu, Biocenter Oulu; University Hospital Oulu; Biocentrum Helsinki; Erasmus Medical Center and Erasmus University, Rotterdam; Karolinska Institutet; Stockholm County Council; Municipality of Rotterdam; Federal State of Mecklenburg-West Pomerania; AstraZeneca; GlaxoSmithKline; Siemens Healthcare; Novo Nordisk Foundation; Yrjö Jahnsson Foundation; Biomedicum Helsinki Foundation; Gyllenberg Foundation; Knut and Alice Wallenberg Foundation; Torsten and Ragnar Söderberg Foundation; Robert Dawson Evans Endowment, Boston University School of Medicine; Instrumentarium Science Foundation; Jenny and Antti Wihuri Foundation and the Canadian Primary Biliary Cirrhosis Society. A full list of acknowledgments is provided in the Supplementary Note.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

Study organization and manuscript preparation was done by J.C.C., W.Z., J. Sehmi, X.L., M.N.W., P.V.d.H., H.H., S.S., M.K., M.A.-K., K.S., P.V., H.V., E.E.S., J. Scott, M.-R.J., P.E. and J.S.K. All authors reviewed and had the opportunity to comment on the manuscript. Data collection and analysis in the participating GWASs were done by G.W.M., J.B.W. and N.G.M. (Australian Twin Cohort); C.W., M.C., M.J.B. and P.B.M. (BRIGHT); D.M.W., G. Waeber, P.M.V., P.V., V.M. and X.Y. (CoLaus); D.F.G., G.I.E., G.T., H.H., I.O., K.S. and U.T. (deCODE); J.L., N.G.F., N.J.W. and R.J.F.L. (Fenland); K.H.P. (Finnish Twin Cohort); C.J.O., C.S.F., J.P.L., L.D.A., N.L.H.-C., R.S.V., T.J.L. and W.G. (Framingham Heart Study); A.D., B.K., C.G., C.M. and H.-E.W. (KORA); B.H.R.W., I.M.L., I.P.K., M.M.V.d.K., P.V.d.H. and R.P.S. (LIFELINES); D.D., G.D., H.C.T., I.P., J.C.C., J. Scott, J. Sehmi, J.S.K., M.I.M., P.E., P.F., S.S.-C., W.Z., X.L. and Y.L. (LOLIPOP); B.P.P., B.W.P., B.Z.A., H.S., J.H.S. and V.L. (NESDA); D.I.B., E.J.C.d.G., G. Willemsen, J.-J.H. (Netherlands Twins Register); A.-L.H., A.P., A.R., E.H., M.-R.J. and P.F.O. (Northern Finland Birth Cohort 1966); H. Watkins, J.F.P., M.F. and U.S. (PROCARDIS); A.G.U., A.H., C.M.v.D., H.L.A.J., J.C.M.W., J.N.L.S. and M.K. (Rotterdam Study 1); D.S., F.C., G.R.A., M.U., S.L. and S.S. (SardiNIA); G.H., H.V., H. Wallaschofski, J.P.K., M.M.L., N.F., R.P. and S.E.B. (SHIP); K.R.A., N.R. and T.D.S. (TwinsUK). Biologic associations of loci and bioinformatics were carried out by G.D., W.T., K. Matsuda, V.K., Y.N. and by G.S., L.J.C., P.C. (AlcGen Consortium), C.X., G.M.H., K.A.S. (Canadian Primary Biliary Cirrhosis Consortium), K. Musunuru, T.M.T. (Global Lipids Consortium), E.K.S., I.B.B., L.M.Y.A., T.B.H. (GOLD consortium) and G.E. and T.J. (ICBP-GWAS). Gene expression analyses were done by E.E.S., A.L.D., H.H., G.T., L.L., M.F.M., M.L., S.H. and W.O.C. Metabonomic analyses were done by A.J.K., M.A.-K., M.J.S., P.S., P.W. and T.T. Structural biology was done by M.J.E.S. and M.N.W.

Corresponding authors

Correspondence to John C Chambers, Paul Elliott or Jaspal S Kooner.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A full list of members is given in Supplementary Note.

A full list of members is given in Supplementary Note.

A full list of members is given in Supplementary Note.

A full list of members is given in Supplementary Note.

A full list of members is given in Supplementary Note.

A full list of members is given in Supplementary Note.

A full list of members is given in Supplementary Note.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–20 and Supplementary Figures 1–6 and Supplementary Note. (PDF 5946 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chambers, J., Zhang, W., Sehmi, J. et al. Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma. Nat Genet 43, 1131–1138 (2011). https://doi.org/10.1038/ng.970

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.970

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