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:

Rare MTNR1B variants impairing melatonin receptor 1B function contribute to type 2 diabetes

Abstract

Genome-wide association studies have revealed that common noncoding variants in MTNR1B (encoding melatonin receptor 1B, also known as MT2) increase type 2 diabetes (T2D) risk1,2. Although the strongest association signal was highly significant (P < 1 × 10−20), its contribution to T2D risk was modest (odds ratio (OR) of 1.10–1.15)1,2,3. We performed large-scale exon resequencing in 7,632 Europeans, including 2,186 individuals with T2D, and identified 40 nonsynonymous variants, including 36 very rare variants (minor allele frequency (MAF) <0.1%), associated with T2D (OR = 3.31, 95% confidence interval (CI) = 1.78–6.18; P = 1.64 × 10−4). A four-tiered functional investigation of all 40 mutants revealed that 14 were non-functional and rare (MAF < 1%), and 4 were very rare with complete loss of melatonin binding and signaling capabilities. Among the very rare variants, the partial- or total-loss-of-function variants but not the neutral ones contributed to T2D (OR = 5.67, CI = 2.17–14.82; P = 4.09 × 10−4). Genotyping the four complete loss-of-function variants in 11,854 additional individuals revealed their association with T2D risk (8,153 individuals with T2D and 10,100 controls; OR = 3.88, CI = 1.49–10.07; P = 5.37 × 10−3). This study establishes a firm functional link between MTNR1B and T2D risk.

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: Distribution of the 40 nonsynonymous MT2 variants identified by exon resequencing.
Figure 2: Functional characterization of wild-type and mutant MT2 receptors.
Figure 3: Odds ratio estimates of partial or total loss-of-function variants compared to neutral very rare (MAF <0.1%) variants for T2D risk determined on the basis of MTNR1B sequencing data.

Similar content being viewed by others

References

  1. Bouatia-Naji, N. et al. A variant near MTNR1B is associated with increased fasting plasma glucose levels and type 2 diabetes risk. Nat. Genet. 41, 89–94 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Prokopenko, I. et al. Variants in MTNR1B influence fasting glucose levels. Nat. Genet. 41, 77–81 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. 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  PubMed  PubMed Central  Google Scholar 

  4. Bass, J. & Takahashi, J.S. Circadian integration of metabolism and energetics. Science 330, 1349–1354 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Marcheva, B. et al. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466, 627–631 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Dubocovich, M.L. et al. International Union of Basic and Clinical Pharmacology. LXXV. Nomenclature, classification, and pharmacology of G protein–coupled melatonin receptors. Pharmacol. Rev. 62, 343–380 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jockers, R., Maurice, P., Boutin, J.A. & Delagrange, P. Melatonin receptors, heterodimerization, signal transduction and binding sites: what′s new? Br. J. Pharmacol. 154, 1182–1195 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Manolio, T.A. et al. Finding the missing heritability of complex diseases. Nature 461, 747–753 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schork, N.J., Murray, S.S., Frazer, K.A. & Topol, E.J. Common vs. rare allele hypotheses for complex diseases. Curr. Opin. Genet. Dev. 19, 212–219 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nejentsev, S., Walker, N., Riches, D., Egholm, M. & Todd, J.A. Rare variants of IFIH1, a gene implicated in antiviral responses, protect against type 1 diabetes. Science 324, 387–389 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Downes, K. et al. Reduced expression of IFIH1 is protective for type 1 diabetes. PLoS ONE 5, e12646 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Johansen, C.T. et al. Excess of rare variants in genes identified by genome-wide association study of hypertriglyceridemia. Nat. Genet. 42, 684–687 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Coventry, A. et al. Deep resequencing reveals excess rare recent variants consistent with explosive population growth. Nat. Commun. 1, 131 (2010).

    Article  PubMed  Google Scholar 

  14. Liu, D.J. & Leal, S.M. A novel adaptive method for the analysis of next-generation sequencing data to detect complex trait associations with rare variants due to gene main effects and interactions. PLoS Genet. 6, e1001156 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Nygaard, R., Frimurer, T.M., Holst, B., Rosenkilde, M.M. & Schwartz, T.W. Ligand binding and micro-switches in 7TM receptor structures. Trends Pharmacol. Sci. 30, 249–259 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Adzhubei, I.A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Andersson, E.A. et al. MTNR1B G24E variant associates with BMI and fasting plasma glucose in the general population in studies of 22,142 Europeans. Diabetes 59, 1539–1548 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lyssenko, V. et al. Common variant in MTNR1B associated with increased risk of type 2 diabetes and impaired early insulin secretion. Nat. Genet. 41, 82–88 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Peschke, E. & Muhlbauer, E. New evidence for a role of melatonin in glucose regulation. Best Pract. Res. Clin. Endocrinol. Metab. 24, 829–841 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Dubovsky, S.L. & Warren, C. Agomelatine, a melatonin agonist with antidepressant properties. Expert Opin. Investig. Drugs 18, 1533–1540 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Balkau, B. An epidemiologic survey from a network of French Health Examination Centres (D.E.S.I.R.): epidemiologic data on the insulin resistance syndrome. Rev. Epidemiol. Sante Publique 44, 373–375 (1996).

    CAS  PubMed  Google Scholar 

  22. Williams, D.R. et al. Undiagnosed glucose intolerance in the community: the Isle of Ely Diabetes Project. Diabet. Med. 12, 30–35 (1995).

    Article  PubMed  Google Scholar 

  23. Syddall, H.E. et al. Cohort profile: the Hertfordshire cohort study. Int. J. Epidemiol. 34, 1234–1242 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Meyre, D. et al. A genome-wide scan for childhood obesity-associated traits in French families shows significant linkage on chromosome 6q22.31-q23.2. Diabetes 53, 803–811 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Hercberg, S. et al. A primary prevention trial using nutritional doses of antioxidant vitamins and minerals in cardiovascular diseases and cancers in a general population: the SU.VI.MAX study—design, methods, and participant characteristics. Supplementation en VItamines et Mineraux Antioxydants. Control. Clin. Trials 19, 336–351 (1998).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  27. Hadjadj, S. et al. Prognostic value of the insertion/deletion polymorphism of the ACE gene in type 2 diabetic subjects: results from the non-insulin-dependent Diabetes, Hypertension, Microalbuminuria or Proteinuria, Cardiovascular Events, and Ramipril (DIABHYCAR), Diabete de type 2, Nephropathie et Genetique (DIAB2NEPHROGENE), and Survie, Diabete de type 2 et Genetique (SURDIAGENE) studies. Diabetes Care 31, 1847–1852 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. American Diabetes Association. Standards of medical care in diabetes. Diabetes Care 31, (suppl.), S12–S54 (2008).

  29. Ayoub, M.A. et al. Monitoring of ligand-independent dimerization and ligand-induced conformational changes of melatonin receptors in living cells by bioluminescence resonance energy transfer. J. Biol. Chem. 277, 21522–21528 (2002).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are sincerely indebted to all participants in the genetic study. We thank M. Deweirder and F. Allegaert for their technical assistance and their invaluable management of DNA samples and B. Gardiola-Lemaître for her invaluable advice. This study was supported by the French Agence Nationale de la Recherche (ANR-08-GENOPAT to P.F. and ANR-11-blanc 'MLT2D' and ANR-11-META 'MELA-BETES' to R.J. and P.F.), the Contrat de Projets Etat-Région Nord-Pas-De-Calais (CPER 2007-2013 'Axe Cardio-Diabète' to P.F.), the Société Francophone du Diabète (to A.B.), the Fondation Recherche Médicale ('Equipe FRM' to R.J.), Inserm and CNRS. I.B. acknowledges funding from the Wellcome Trust (077016/Z/05/Z) and from the UK National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre and the MRC Centre for Obesity and Related Metabolic Diseases.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

A.B., R.J. and P.F. wrote the manuscript, and N.C., J.-L.G., M.V., C.D., R.S., M.I.M. and I.B. reviewed and/or edited the manuscript and contributed to discussion. R.J. and P.F. managed the project. L.Y. performed the statistical analyses, and A.B., G.R. and C.D. contributed to statistical analyses. A.B. and K.F. performed the sequencing. E.V. performed the genotyping. A.B., N.C., E.V., J.-L.G. and A.D. performed the functional analyses. F.P., R.R., S.C., S. Hercberg, S. Hadjadj, B.B., M.M., O.L., C.L., N.B.-N., MAGIC, G.C., N.J.W., M.I.M. and I.B. contributed to cohort study samples and researched data.

Corresponding authors

Correspondence to Ralf Jockers or Philippe Froguel.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A list of members is provided in the Supplementary Note.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–4, Supplementary Figures 1–4 and Supplementary Note (PDF 707 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bonnefond, A., Clément, N., Fawcett, K. et al. Rare MTNR1B variants impairing melatonin receptor 1B function contribute to type 2 diabetes. Nat Genet 44, 297–301 (2012). https://doi.org/10.1038/ng.1053

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.1053

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