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:

Assessing the phenotypic effects in the general population of rare variants in genes for a dominant Mendelian form of diabetes

Abstract

Genome sequencing can identify individuals in the general population who harbor rare coding variants in genes for Mendelian disorders1,2,3,4,5,6,7 and who may consequently have increased disease risk. Previous studies of rare variants in phenotypically extreme individuals display ascertainment bias and may demonstrate inflated effect-size estimates8,9,10,11,12. We sequenced seven genes for maturity-onset diabetes of the young (MODY)13 in well-phenotyped population samples14,15 (n = 4,003). We filtered rare variants according to two prediction criteria for disease-causing mutations: reported previously in MODY or satisfying stringent de novo thresholds (rare, conserved and protein damaging). Approximately 1.5% and 0.5% of randomly selected individuals from the Framingham and Jackson Heart Studies, respectively, carry variants from these two classes. However, the vast majority of carriers remain euglycemic through middle age. Accurate estimates of variant effect sizes from population-based sequencing are needed to avoid falsely predicting a substantial fraction of individuals as being at risk for MODY or other Mendelian diseases.

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: Description of low frequency nonsynonymous variants.
Figure 2: Phenotypic impact of variants in the unselected cohorts.
Figure 3: Phenotypes of GCK and HNF1A variant carriers.

Similar content being viewed by others

Accession codes

Primary accessions

NCBI Reference Sequence

Referenced accessions

NCBI Reference Sequence

References

  1. Collins, F.S. Shattuck lecture—medical and societal consequences of the Human Genome Project. N. Engl. J. Med. 341, 28–37 (1999).

    Article  CAS  PubMed  Google Scholar 

  2. Collins, F.S. Genetics: an explosion of knowledge is transforming clinical practice. Geriatrics 54, 41–47, quiz 48 (1999).

    CAS  PubMed  Google Scholar 

  3. Roses, A.D. Pharmacogenetics and the practice of medicine. Nature 405, 857–865 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Hall, Y. Coming Soon: Your Personal DNA Map? National Geographic Newshttp://news.nationalgeographic.com/news/2006/03/0307_060307_dna.html〉 (2006).

  5. Duncan, D.E. On a Mission to Sequence the Genomes of 100,000 People. New York Times (7 June 2010).

  6. Brunham, L.R. & Hayden, M.R. Medicine. Whole-genome sequencing: the new standard of care? Science 336, 1112–1113 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Ball, M.P. et al. A public resource facilitating clinical use of genomes. Proc. Natl. Acad. Sci. USA 109, 11920–11927 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Begg, C.B. On the use of familial aggregation in population-based case probands for calculating penetrance. J. Natl. Cancer Inst. 94, 1221–1226 (2002).

    Article  PubMed  Google Scholar 

  9. Beutler, E., Felitti, V.J., Koziol, J.A., Ho, N.J. & Gelbart, T. Penetrance of 845G→A (C282Y) HFE hereditary haemochromatosis mutation in the USA. Lancet 359, 211–218 (2002).

    Article  PubMed  Google Scholar 

  10. Göring, H.H., Terwilliger, J.D. & Blangero, J. Large upward bias in estimation of locus-specific effects from genomewide scans. Am. J. Hum. Genet. 69, 1357–1369 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Guey, L.T. et al. Power in the phenotypic extremes: a simulation study of power in discovery and replication of rare variants. Genet. Epidemiol. 35, 236–246 (2011).

    PubMed  Google Scholar 

  12. Terwilliger, J.D. & Weiss, K.M. Confounding, ascertainment bias, and the blind quest for a genetic 'fountain of youth'. Ann. Med. 35, 532–544 (2003).

    Article  PubMed  Google Scholar 

  13. Molven, A. & Njolstad, P.R. Role of molecular genetics in transforming diagnosis of diabetes mellitus. Expert Rev. Mol. Diagn. 11, 313–320 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Kannel, W.B., Feinleib, M., McNamara, P.M., Garrison, R.J. & Castelli, W.P. An investigation of coronary heart disease in families. The Framingham offspring study. Am. J. Epidemiol. 110, 281–290 (1979).

    Article  CAS  PubMed  Google Scholar 

  15. Sempos, C.T., Bild, D.E. & Manolio, T.A. Overview of the Jackson Heart Study: a study of cardiovascular diseases in African American men and women. Am. J. Med. Sci. 317, 142–146 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Begg, C.B. et al. Variation of breast cancer risk among BRCA1/2 carriers. J. Am. Med. Assoc. 299, 194–201 (2008).

    Article  CAS  Google Scholar 

  17. Kohane, I.S., Hsing, M. & Kong, S.W. Taxonomizing, sizing, and overcoming the incidentalome. Genet. Med. 14, 399–404 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Tattersall, R.B. Mild familial diabetes with dominant inheritance. Q. J. Med. 43, 339–357 (1974).

    CAS  PubMed  Google Scholar 

  19. Tattersall, R.B. & Fajans, S.S. A difference between the inheritance of classical juvenile-onset and maturity-onset type diabetes of young people. Diabetes 24, 44–53 (1975).

    Article  CAS  PubMed  Google Scholar 

  20. Murphy, R., Ellard, S. & Hattersley, A.T. Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes. Nat. Clin. Pract. Endocrinol. Metab. 4, 200–213 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Eide, S.A. et al. Prevalence of HNF1A (MODY3) mutations in a Norwegian population (the HUNT2 Study). Diabet. Med. 25, 775–781 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Ledermann, H.M. Is maturity onset diabetes at young age (MODY) more common in Europe than previously assumed? Lancet 345, 648 (1995).

    Article  CAS  PubMed  Google Scholar 

  23. Shields, B.M. et al. Maturity-onset diabetes of the young (MODY): how many cases are we missing? Diabetologia 53, 2504–2508 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Shepherd, M. et al. No deterioration in glycemic control in HNF-1α maturity-onset diabetes of the young following transfer from long-term insulin to sulphonylureas. Diabetes Care 26, 3191–3192 (2003).

    Article  PubMed  Google Scholar 

  25. Shepherd, M. & Hattersley, A.T. 'I don't feel like a diabetic any more': the impact of stopping insulin in patients with maturity onset diabetes of the young following genetic testing. Clin. Med. (Northfield IL) 4, 144–147 (2004).

    Article  Google Scholar 

  26. Shepherd, M. et al. Predictive genetic testing in maturity-onset diabetes of the young (MODY). Diabet. Med. 18, 417–421 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. McCarthy, M.I. Genomics, type 2 diabetes, and obesity. N. Engl. J. Med. 363, 2339–2350 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Knowler, W.C. et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N. Engl. J. Med. 346, 393–403 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Knowler, W.C. et al. 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet 374, 1677–1686 (2009).

    Article  PubMed  Google Scholar 

  30. Yamagata, K. et al. Mutations in the hepatocyte nuclear factor-1α gene in maturity-onset diabetes of the young (MODY3). Nature 384, 455–458 (1996).

    Article  CAS  PubMed  Google Scholar 

  31. Hattersley, A.T. et al. Linkage of type 2 diabetes to the glucokinase gene. Lancet 339, 1307–1310 (1992).

    Article  CAS  PubMed  Google Scholar 

  32. Froguel, P. et al. Familial hyperglycemia due to mutations in glucokinase. Definition of a subtype of diabetes mellitus. N. Engl. J. Med. 328, 697–702 (1993).

    Article  CAS  PubMed  Google Scholar 

  33. Yamagata, K. et al. Mutations in the hepatocyte nuclear factor-4α gene in maturity-onset diabetes of the young (MODY1). Nature 384, 458–460 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Horikawa, Y. et al. Mutation in hepatocyte nuclear factor-1β gene (TCF2) associated with MODY. Nat. Genet. 17, 384–385 (1997).

    Article  CAS  PubMed  Google Scholar 

  35. Stoffers, D.A., Ferrer, J., Clarke, W.L. & Habener, J.F. Early-onset type-II diabetes mellitus (MODY4) linked to IPF1. Nat. Genet. 17, 138–139 (1997).

    Article  CAS  PubMed  Google Scholar 

  36. Molven, A. et al. Mutations in the insulin gene can cause MODY and autoantibody-negative type 1 diabetes. Diabetes 57, 1131–1135 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Malecki, M.T. et al. Mutations in NEUROD1 are associated with the development of type 2 diabetes mellitus. Nat. Genet. 23, 323–328 (1999).

    Article  CAS  PubMed  Google Scholar 

  38. Flanagan, S.E. et al. Update of mutations in the genes encoding the pancreatic beta-cell K (ATP) channel subunits Kir6.2 (KCNJ11) and sulfonylurea receptor 1 (ABCC8) in diabetes mellitus and hyperinsulinism. Hum. Mutat. 30, 170–180 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Plengvidhya, N. et al. PAX4 mutations in Thais with maturity onset diabetes of the young. J. Clin. Endocrinol. Metab. 92, 2821–2826 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Borowiec, M. et al. Mutations at the BLK locus linked to maturity onset diabetes of the young and beta-cell dysfunction. Proc. Natl. Acad. Sci. USA 106, 14460–14465 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Neve, B. et al. Role of transcription factor KLF11 and its diabetes-associated gene variants in pancreatic beta cell function. Proc. Natl. Acad. Sci. USA 102, 4807–4812 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Raeder, H. et al. Mutations in the CEL VNTR cause a syndrome of diabetes and pancreatic exocrine dysfunction. Nat. Genet. 38, 54–62 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Gnirke, A. et al. Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing. Nat. Biotechnol. 27, 182–189 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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  PubMed  PubMed Central  Google Scholar 

  45. 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 

  46. Abecasis, G.R. et al. A map of human genome variation from population-scale sequencing. Nature 467, 1061–1073 (2010).

    Article  PubMed  CAS  Google Scholar 

  47. Kumar, P., Henikoff, S. & Ng, P.C. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 4, 1073–1081 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. 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 

  49. Stenson, P.D. et al. The Human Gene Mutation Database: 2008 update. Genome Med. 1, 13 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Xue, Y. et al. Deleterious- and disease-allele prevalence in healthy individuals: insights from current predictions, mutation databases, and population-scale resequencing. Am. J. Hum. Genet. 91, 1022–1032 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. MacArthur, D.G. et al. A systematic survey of loss-of-function variants in human protein-coding genes. Science 335, 823–828 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Srinivasan, B.S. et al. A universal carrier test for the long tail of Mendelian disease. Reprod. Biomed. Online 21, 537–551 (2010).

    Article  PubMed  Google Scholar 

  53. Arthur, C. Mapping the IndividualCheaply. The Guardian (23 April 2008).

  54. Pinker, S. My Genome, My Self. New York Times (7 January 2009).

  55. Rochman, B. The DNA dilemma: a test that could change your life. TIME Magazine (2012).

  56. Lango Allen, H. et al. GATA6 haploinsufficiency causes pancreatic agenesis in humans. Nat. Genet. 44, 20–22 (2012).

    Article  CAS  Google Scholar 

  57. Johansson, S. et al. Exome sequencing and genetic testing for MODY. PLoS ONE 7, e38050 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Eide, S.A. et al. Prevalence of HNF1A (MODY3) mutations in a Norwegian population (the HUNT2 Study). Diabetic Med. 25, 775–81 (2008).

    Article  CAS  PubMed  Google Scholar 

  59. Ellard, S., Bellanne-Chantelot, C. & Hattersley, A.T. Best practice guidelines for the molecular genetic diagnosis of maturity-onset diabetes of the young. Diabetologia 51, 546–553 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. World Health Organization. Definition and Diagnosis of Diabetes Mellitus and Intermediate Hyperglycaemia: Report of a WHO/IDF Consultation (World Health Organization Press, Geneva, 2006).

  61. Edghill, E.L. et al. Sequencing PDX1 (insulin promoter factor 1) in 1788 UK individuals found 5% had a low frequency coding variant, but these variants are not associated with Type 2 diabetes. Diabet. Med. 28, 681–684 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Gill-Carey, O., Shields, B., Colclough, K., Ellard, S. & Hattersley, A. Finding a glucokinase mutation alters patient treatment. Diabetic Med. 24 (suppl. 1), 6 (2007).

  63. Pearson, E.R. et al. Beta-cell genes and diabetes: quantitative and qualitative differences in the pathophysiology of hepatic nuclear factor-1α and glucokinase mutations. Diabetes 50 (suppl. 1), S101–S107 (2001).

    Article  CAS  PubMed  Google Scholar 

  64. Stride, A. et al. The genetic abnormality in the beta cell determines the response to an oral glucose load. Diabetologia 45, 427–435 (2002).

    Article  CAS  PubMed  Google Scholar 

  65. Bergmann, A. et al. The A98V single nucleotide polymorphism (SNP) in hepatic nuclear factor 1α (HNF-1α) is associated with insulin sensitivity and beta-cell function. Exp. Clin. Endocrinol. Diabetes 116 (suppl. 1), S50–S55 (2008).

    Article  CAS  PubMed  Google Scholar 

  66. Iwasaki, N. et al. Liver and kidney function in Japanese patients with maturity-onset diabetes of the young. Diabetes Care 21, 2144–2148 (1998).

    Article  CAS  PubMed  Google Scholar 

  67. McLaren, W. et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26, 2069–2070 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Bach, I. et al. Cloning of human hepatic nuclear factor 1 (HNF1) and chromosomal localization of its gene in man and mouse. Genomics 8, 155–164 (1990).

    Article  CAS  PubMed  Google Scholar 

  69. Chartier, F.L., Bossu, J.P., Laudet, V., Fruchart, J.C. & Laine, B. Cloning and sequencing of cDNAs encoding the human hepatocyte nuclear factor 4 indicate the presence of two isoforms in human liver. Gene 147, 269–272 (1994).

    Article  CAS  PubMed  Google Scholar 

  70. Abbott, C. et al. Mapping of the gene TCF2 coding for the transcription factor LFB3 to human chromosome 17 by polymerase chain reaction. Genomics 8, 165–167 (1990).

    Article  CAS  PubMed  Google Scholar 

  71. Leonard, J. et al. Characterization of somatostatin transactivating factor-1, a novel homeobox factor that stimulates somatostatin expression in pancreatic islet cells. Mol. Endocrinol. 7, 1275–1283 (1993).

    CAS  PubMed  Google Scholar 

  72. Sanger, F. Chemistry of insulin; determination of the structure of insulin opens the way to greater understanding of life processes. Science 129, 1340–1344 (1959).

    Article  CAS  PubMed  Google Scholar 

  73. Tamimi, R. et al. The NEUROD gene maps to human chromosome 2q32 and mouse chromosome 2. Genomics 34, 418–421 (1996).

    Article  CAS  PubMed  Google Scholar 

  74. Dawber, T.R., Meadors, G.F. & Moore, F.E. Jr. Epidemiological approaches to heart disease: the Framingham Study. Am. J. Public Health Nations Health 41, 279–281 (1951).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Berglund, G. et al. Long-term outcome of the Malmo preventive project: mortality and cardiovascular morbidity. J. Intern. Med. 247, 19–29 (2000).

    Article  CAS  PubMed  Google Scholar 

  76. Lindholm, E., Agardh, E., Tuomi, T., Groop, L. & Agardh, C.D. Classifying diabetes according to the new WHO clinical stages. Eur. J. Epidemiol. 17, 983–989 (2001).

    Article  CAS  PubMed  Google Scholar 

  77. Groop, L. et al. Metabolic consequences of a family history of NIDDM (the Botnia study): evidence for sex-specific parental effects. Diabetes 45, 1585–1593 (1996).

    Article  CAS  PubMed  Google Scholar 

  78. Byrne, M.M. et al. Altered insulin secretory responses to glucose in diabetic and nondiabetic subjects with mutations in the diabetes susceptibility gene MODY3 on chromosome 12. Diabetes 45, 1503–1510 (1996).

    Article  CAS  PubMed  Google Scholar 

  79. Bick, A.G. et al. Burden of rare sarcomere gene variants in the Framingham and Jackson Heart Study cohorts. Am. J. Hum. Genet. 91, 513–519 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. DePristo, M.A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  85. Meyer, L.R. et al. The UCSC Genome Browser database: extensions and updates 2013. Nucleic Acids Res. 41, D64–D69 (2013).

    Article  CAS  PubMed  Google Scholar 

  86. Lindner, T.H. et al. A novel syndrome of diabetes mellitus, renal dysfunction and genital malformation associated with a partial deletion of the pseudo-POU domain of hepatocyte nuclear factor-1β. Hum. Mol. Genet. 8, 2001–2008 (1999).

    Article  CAS  PubMed  Google Scholar 

  87. Njølstad, P.R. et al. Permanent neonatal diabetes caused by glucokinase deficiency: inborn error of the glucose-insulin signaling pathway. Diabetes 52, 2854–2860 (2003).

    Article  PubMed  Google Scholar 

  88. Bjørkhaug, L. et al. Hepatocyte nuclear factor-1α gene mutations and diabetes in Norway. J. Clin. Endocrinol. Metab. 88, 920–931 (2003).

    Article  PubMed  CAS  Google Scholar 

  89. Sagen, J.V. et al. Diagnostic screening of NEUROD1 (MODY6) in subjects with MODY or gestational diabetes mellitus. Diabetic Med. 22, 1012–1015 (2005).

    Article  CAS  PubMed  Google Scholar 

  90. Raeder, H. et al. A hepatocyte nuclear factor-4α gene (HNF4A) P2 promoter haplotype linked with late-onset diabetes: studies of HNF4A variants in the Norwegian MODY registry. Diabetes 55, 1899–1903 (2006).

    Article  CAS  PubMed  Google Scholar 

  91. Sagen, J.V. et al. From clinicogenetic studies of maturity-onset diabetes of the young to unraveling complex mechanisms of glucokinase regulation. Diabetes 55, 1713–1722 (2006).

    Article  CAS  PubMed  Google Scholar 

  92. Haldorsen, I.S. et al. Lack of pancreatic body and tail in HNF1B mutation carriers. Diabetic Med. 25, 782–787 (2008).

    Article  CAS  PubMed  Google Scholar 

  93. Sagen, J.V. et al. Diagnostic screening of MODY2/GCK mutations in the Norwegian MODY Registry. Pediatr. Diabetes 9, 442–449 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge the contribution of the participants of the Framingham and Jackson Heart Study, as well as the participants from the Malmö Preventive Project, the Scania Diabetes Registry and the Botnia Study. This work was supported by grants from the National Human Genome Research Institute of the US National Institutes of Health (NIH) (Medical Sequencing Program grant U54 HG003067 to the Broad Institute principal investigator, E. Lander) and the Howard Hughes Medical Institute, as well as funding from Pfizer Inc. J.F. was supported in part by NIH training grant 5-T32-GM007748-33. N.L.B. was supported by a Fulbright Diabetes UK Fellowship (BDA 11/0004348). D.A. was supported by funding from the Doris Duke Charitable Foundation (2006087). J.M. acknowledges support from National Institute of Diabetes and Digestive and Kidney Diseases grant K24 DK080140. J.B.M. and J.C.F. acknowledge support from NIH grant R01 DK078616. A.G.B. and V.A. were supported by NIH Medical Scientist Training Program fellowship T32GM007753. J.G.S. and C.E.S. were supported by NIH RO1 2R01HL080494, the National Heart, Lung, and Blood Institute (NHLBI) and the LeDucq Foundation. The Jackson Heart Study is supported by contracts N01-HC-95170, N01-HC-95171 and N01-HC-95172 from the NHLBI, the National Institute for Minority Health and Health Disparities and additional support from the National Institute of Biomedical Imaging and Bioengineering. The Framingham Heart Study was supported by contracts N01-HC-25195 and 6R01-NS 17950 from the NHLBI and genotyping services from Affymetrix, Inc. (contract N02-HL-6-4278 for the SNP Health Association Resource, SHARe, project). The Malmö Preventive Project and the Scania Diabetes Registry were supported by a Swedish Research Council grant (Linné) to the Lund University Diabetes Centre. The Botnia study was supported by funding from the Sigrid Juselius Foundation and the Folkhälsan Research Foundation, as well as a European Research Council advanced research grant to L.G. (GA 269045). The MODY study was supported by grants from the KG Jebsen Foundation, the Norwegian Research Council, the University of Bergen, Helse Vest, Innovest and the European Research Council (AdG).

Author information

Authors and Affiliations

Authors

Contributions

This manuscript describes an integrated analysis that draws together two studies that were initially independent: the Pfizer/Massachusetts General Hospital/Broad/Lund collaborative project 'Towards Therapeutic Targets for Type 2 Diabetes and Myocardial Infarction in the Background of Type 2 Diabetes' (PMBL), which was supervised by D.A., L.G., T.R., D.R.C., S.K., J.K.T. and M.J.B., and the National Human Genome Research Institute–funded project 'Analyses of the Allelic Spectrum of Cardiovascular Disease Genes in the Framingham Heart Study and Jackson Heart Study Cohorts' (FHS/JHS), which was led by C.E.S. and S.G. For the PMBL project, N.P.B. was the project manager, and L.G., T.T., V.L. and F.B. were responsible for clinical investigation and sample management. For the FHS/JHS project, N.G. was the project manager; H.T., E.F., J.G.W. and C.J.O. were responsible for clinical investigation and sample management; and J.G.W., C.J.O., C.N.-C., S.K., J.N.H., J.G.S., D.A., S.G. and C.E.S. supervised the project. All DNA sequencing and data processing for these two projects was performed at the Broad Institute. J.M., H.I., S.J., A.M. and P.R.N. were responsible for all clinical investigation, sample management, sequencing and data processing for the MODY study. J.F., N.L.B. and D.A. designed and conceived the joint analysis. J.F., N.L.B., J.C.F., J.B.M. and D.A. provided methodological expertise. N.L.B., A.G.B., J.F. and V.A. defined and interpreted the clinical information included. J.F. performed all analyses of the three population-based cohorts and performed annotation and comparative analysis of the patients from the MODY study. J.F., N.L.B. and D.A. wrote the manuscript. All authors reviewed, edited and approved the manuscript.

Corresponding author

Correspondence to David Altshuler.

Ethics declarations

Competing interests

M.J.B., J.K.T. and T.R. are employees of Pfizer, Inc. F.B. and D.R.C. are former employees of Pfizer, Inc.; F.B. retains shares in the company.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Tables 1–10 (PDF 1792 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Flannick, J., Beer, N., Bick, A. et al. Assessing the phenotypic effects in the general population of rare variants in genes for a dominant Mendelian form of diabetes. Nat Genet 45, 1380–1385 (2013). https://doi.org/10.1038/ng.2794

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2794

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