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:

Variants in MTNR1B influence fasting glucose levels

Abstract

To identify previously unknown genetic loci associated with fasting glucose concentrations, we examined the leading association signals in ten genome-wide association scans involving a total of 36,610 individuals of European descent. Variants in the gene encoding melatonin receptor 1B (MTNR1B) were consistently associated with fasting glucose across all ten studies. The strongest signal was observed at rs10830963, where each G allele (frequency 0.30 in HapMap CEU) was associated with an increase of 0.07 (95% CI = 0.06–0.08) mmol/l in fasting glucose levels (P = 3.2 × 10−50) and reduced beta-cell function as measured by homeostasis model assessment (HOMA-B, P = 1.1 × 10−15). The same allele was associated with an increased risk of type 2 diabetes (odds ratio = 1.09 (1.05–1.12), per G allele P = 3.3 × 10−7) in a meta-analysis of 13 case-control studies totaling 18,236 cases and 64,453 controls. Our analyses also confirm previous associations of fasting glucose with variants at the G6PC2 (rs560887, P = 1.1 × 10−57) and GCK (rs4607517, P = 1.0 × 10−25) loci.

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

Access options

Buy this article

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

Figure 1: Regional plot of fasting glucose association results for the MTNR1B locus across ten MAGIC GWAS.
Figure 2: Association of rs10830963 with type 2 diabetes (T2D) in 13 case-control studies.

Similar content being viewed by others

References

  1. Xiang, A.H. et al. Coordinate changes in plasma glucose and pancreatic beta-cell function in Latino women at high risk for type 2 diabetes. Diabetes 55, 1074–1079 (2006).

    Article  CAS  Google Scholar 

  2. Mason, C.C., Hanson, R.L. & Knowler, W.C. Progression to type 2 diabetes characterized by moderate then rapid glucose increases. Diabetes 56, 2054–2061 (2007).

    Article  CAS  Google Scholar 

  3. Watanabe, R.M. et al. Familiality of quantitative metabolic traits in Finnish families with non-insulin-dependent diabetes mellitus. Finland-United States Investigation of NIDDM Genetics (FUSION) Study investigators. Hum. Hered. 49, 159–168 (1999).

    Article  CAS  Google Scholar 

  4. Weedon, M.N. et al. A common haplotype of the glucokinase gene alters fasting glucose and birth weight: association in six studies and population-genetics analyses. Am. J. Hum. Genet. 79, 991–1001 (2006).

    Article  CAS  Google Scholar 

  5. Bouatia-Naji, N. et al. A polymorphism within the G6PC2 gene is associated with fasting plasma glucose levels. Science 320, 1085–1088 (2008).

    Article  CAS  Google Scholar 

  6. Chen, W.M. et al. Variations in the G6PC2/ABCB11 genomic region are associated with fasting glucose levels. J. Clin. Invest. 118, 2620–2628 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Vaxillaire, M. et al. Impact of common type 2 diabetes risk polymorphisms in the DESIR prospective study. Diabetes 57, 244–254 (2008).

    Article  CAS  Google Scholar 

  9. Orho-Melander, M. et al. A common missense variant in the glucokinase regulatory protein gene (GCKR) is associated with increased plasma triglyceride and C-reactive protein but lower fasting glucose concentrations. Diabetes 57, 3112–3121 (2008).

    Article  CAS  Google Scholar 

  10. 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 

  11. Scott, L.J. et al. A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316, 1341–1345 (2007).

    Article  CAS  Google Scholar 

  12. Sladek, R. et al. A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445, 881–885 (2007).

    Article  CAS  Google Scholar 

  13. Steinthorsdottir, V. et al. A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat. Genet. 39, 770–775 (2007).

    Article  CAS  Google Scholar 

  14. Zeggini, E. et al. Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes. Science 316, 1336–1341 (2007).

    Article  CAS  Google Scholar 

  15. The International HapMap Consortium. A haplotype map of the human genome. Nature 437, 1299–1320 (2005).

  16. Matthews, D.R. et al. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28, 412–419 (1985).

    Article  CAS  Google Scholar 

  17. Hofman, A. et al. The Rotterdam Study: objectives and design update. Eur. J. Epidemiol. 22, 819–829 (2007).

    Article  Google Scholar 

  18. Herder, C. et al. Variants of the PPARG, IGF2BP2, CDKAL1, HHEX, and TCF7L2 genes confer risk of type 2 diabetes independently of BMI in the German KORA Studies. Horm. Metab. Res. 40, 722–726 (2008).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  20. Stranger, B.E. et al. Population genomics of human gene expression. Nat. Genet. 39, 1217–1224 (2007).

    Article  CAS  Google Scholar 

  21. Myers, A.J. et al. A survey of genetic human cortical gene expression. Nat. Genet. 39, 1494–1499 (2007).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  23. Reppert, S.M. et al. Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc. Natl. Acad. Sci. USA 92, 8734–8738 (1995).

    Article  CAS  Google Scholar 

  24. Su, A.I. et al. Large-scale analysis of the human and mouse transcriptomes. Proc. Natl. Acad. Sci. USA 99, 4465–4470 (2002).

    Article  CAS  Google Scholar 

  25. Ramracheya, R.D. et al. Function and expression of melatonin receptors on human pancreatic islets. J. Pineal Res. 44, 273–279 (2008).

    Article  CAS  Google Scholar 

  26. Stumpf, I., Muhlbauer, E. & Peschke, E. Involvement of the cGMP pathway in mediating the insulin-inhibitory effect of melatonin in pancreatic beta-cells. J. Pineal Res. 45, 318–327 (2008).

    Article  CAS  Google Scholar 

  27. Boden, G., Ruiz, J., Urbain, J.L. & Chen, X. Evidence for a circadian rhythm of insulin secretion. Am. J. Physiol. 271, E246–E252 (1996).

    CAS  PubMed  Google Scholar 

  28. Spiegel, K., Leproult, R. & Van, C.E. Impact of sleep debt on metabolic and endocrine function. Lancet 354, 1435–1439 (1999).

    Article  CAS  Google Scholar 

  29. Turek, F.W. et al. Obesity and metabolic syndrome in circadian Clock mutant mice. Science 308, 1043–1045 (2005).

    Article  CAS  Google Scholar 

  30. Peschke, E. et al. Melatonin and type 2 diabetes - a possible link? J. Pineal Res. 42, 350–358 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the many colleagues who contributed to collection and phenotypic characterization of the clinical samples, as well as genotyping and analysis of the GWA data. They would also like to acknowledge those who agreed to participate in these studies. Major funding for the work described in this paper comes from Academy of Finland (124243); the Administration of Lanusei, Ilbono, Arzana and Elini (Sardinia, Italy); American Diabetes Association (1-05-RA-140); the Center for Inherited Disease Research; Clinical Research Institute (HUCH); Diabetes UK; the European Bioinformatics Institute; the European Commission (contracts LSHM-CT-2006-037197, LSHM-CT-2003-503041, QLK6-CT-2002-02629, QLG2-CT-2002-01254, HEALTH-F4-2007-201413, LSHG-CT-2004-512066, QLRT-2001-01254, LSHG-CT-2004-518153); the Faculty of Biology and Medicine of Lausanne; Finnish Diabetes Research Foundation; Folkhalsan Research Foundation; Foundation of the NIH (GAIN initiative); German Federal Ministry of Education and Research; German Federal Ministry of Health and Social Security; German National Genome Research Network; GlaxoSmithKline; GSF-National Research Center for Environment and Health; LMUinnovativ; Ministry of Science and Research of the State North-Rhine Westphalia; Municipality of Rotterdam; US National Institutes of Health (HG-02651, HL-084729, HL-087679, HC-25195, N02-HL-6-4278, DK-078616, DK-080140, DK-065978, RR-163736, MH059160, DK069922, DA-021519, DK-062370, DK-072193, US National Human Genome Research Institute intramural project HG-000024; and the Intramural Program of the National Institute on Aging); the UK National Institute for Health Research (Oxford Biomedical Research Centre and Guys and St. Thomas' Biomedical Research Centre); the Netherlands Ministry of Education, Culture and Science; the Netherlands Ministry of Health, Welfare and Sports; Novartis; NWO (904-61-090, 904-61-193, 480-04-004, 400-05-717); NWOGenomics; NWOInvestments; Research Institute for Diseases in the Elderly (RIDE); Sigrid Juselius Foundation; Spinozapremie; Swedish Research Council (349-2006-237); UK Medical Research Council (G0500539, G0000649, G016121); UK National Health Services Research and Development; the Wellcome Trust (including intramural support for the Wellcome Trust Sanger Institute, GR069224, Strategic Awards 076113 and 083948, Biomedical Collections Grant GR072960); and ZonMw (10-000-1002). A full list of acknowledgments is provided in the Supplementary Note.

Author information

Authors and Affiliations

Authors

Contributions

Project management: DFS: R.N.B., A. Cao, F.S.C., K.L.M., J.T., D.S., M.U., E.L., L.C.G., M. Boehnke, G.R.A.; ENGAGE: P.E., A.H., J.H.S., H.-E.W., G. Willemsen, D.I.B., B.W.J.H.P., M.R.J., L.P., U.T., C.v.D., K. Stefansson, M.I.M.; FHS: J.D., J.B.M.; GEM: T.D.S., I.B., N.J.W.

Study design: DFS: R.S., V.L., R.N.B., T.A.B., A. Cao, F.S.C., K.L.M., L.J.S., J.T., D.S., M.U., E.L., M. Boehnke, R.M.W., G.R.A.; ENGAGE: L.P., A.H., U.T., C.v.D., K. Stefansson, M.I.M.; FHS: J.D., J.C.F., J.B.M.; GEM: C.L., N.S., R.J.F.L., J.S.B., M. Bochud, D.W., M.S., T.D.S., P.V., G. Waeber, V.M., I.B., N.J.W.

Genome-wide association sampling and genotyping: DFS: M.R.E., L.L.B., A. Cao, L. Crisponi, T.E.H., B.I., U.L., S.N., M.O., A.S., H.M.S., T.T., J.T., M.U., D.A., L.C.G.; ENGAGE: P.E., N.F., A.P., E.S., V.S., A.G.U., J.C.M.W., D.I.B., M.R.J., L.P., U.T., C.v.D., K. Stefansson, M.I.M.; FHS: C.S.F., L.A.C., J.D., J.B.M.; GEM: K.A., A. Cervino, P.D., M.I., O.T.M.

Statistical analysis and informatics: DFS: R.S., A.U.J., S. Sanna, W.-M.C., V.G., P.S., B.F.V., R.M.W.; ENGAGE: I.P., G.T., Y.A., J.-J.H., M. Kaakinen, L. Coin, E.J.C.d.G., A.D., C.H., A.K., M. Krestyaninova, C.M.L., J.R., V.S., E.Z., C.v.D.; FHS: A.K.M., J.D.; GEM: C.L., N.S., S.C.P., E.W., S.H., T.J., N.L., S. Sharp, K. Song, X.Y., J.H.Z.

Replication sampling and genotyping: A.S.F.D., A.H., T.I., M.L., A.D.M., C.N.A.P., W.R., J.S., H.E.W.

MAGIC management committee: I.P., C.L., J.C.F., R.S., N.S., G.T., R.J.F.L., A.U.J., Y.A., E.W., V.L., C.M.L., D.W., D.S., M.S., P.V., G. Waeber., L.C.G., V.M., U.T., M. Boehnke, I.B., J.D., R.M.W., M.I.M., N.J.W., J.B.M., G.R.A.

Writing team: I.P., C.L., J.C.F., R.S., N.S., G.T., M. Boehnke, I.B., C.v.D., J.D., R.M.W., K. Stefansson, M.I.M., N.J.W., J.B.M., G.R.A.

Corresponding authors

Correspondence to Mark I McCarthy, Nicholas J Wareham, James B Meigs or Gonçalo R Abecasis.

Ethics declarations

Competing interests

Dawn Waterworth and Vincent Mooser are full-time employees of GlaxoSmithKline. Inês Barroso owns stock in GlaxoSmithKline and Incyte. Drs. Thorleifsson, Kong, Steinsthorsdottir, Thorsteinsdottir, and Stefansson are employees at deCODE genetics, and own stock or stock options in the company. Dr. Meigs currently has research grants from GlaxoSmithKline and Sanofi-Aventis, and serves on consultancy boards for GlaxoSmithKline, Sanofi-Aventis, Interleukin Genetics, Kalypsis, and Outcomes Sciences. Dr. Florez has received consulting honoraria from Merck and from Publicis Healthcare Communications, an advertising agency for Amylin Pharmaceuticals.

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Methods, Supplementary Figure 1 and Supplementary Tables 1–6 (PDF 523 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Prokopenko, I., Langenberg, C., Florez, J. et al. Variants in MTNR1B influence fasting glucose levels. Nat Genet 41, 77–81 (2009). https://doi.org/10.1038/ng.290

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.290

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