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.

  • Article
  • Published:

Genetic predisposition for beta cell fragility underlies type 1 and type 2 diabetes

Abstract

Type 1 (T1D) and type 2 (T2D) diabetes share pathophysiological characteristics, yet mechanistic links have remained elusive. T1D results from autoimmune destruction of pancreatic beta cells, whereas beta cell failure in T2D is delayed and progressive. Here we find a new genetic component of diabetes susceptibility in T1D non-obese diabetic (NOD) mice, identifying immune-independent beta cell fragility. Genetic variation in Xrcc4 and Glis3 alters the response of NOD beta cells to unfolded protein stress, enhancing the apoptotic and senescent fates. The same transcriptional relationships were observed in human islets, demonstrating the role of beta cell fragility in genetic predisposition to diabetes.

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: NOD mouse susceptibility to immune-independent diabetes demonstrated through a sensitized transgenic model.
Figure 2: Transgene-induced beta cell stress results in disturbed insulin processing and glucose intolerance.
Figure 3: Qualitative rather than quantitative differences in the UPR on the B10 and NOD backgrounds.
Figure 4: Genetic control of NOD mouse susceptibility to transgene-induced diabetes.
Figure 5: Xrcc4 mutation drives enhanced susceptibility to senescence.
Figure 6: Reduced Glis3 expression results in enhanced susceptibility to apoptosis.
Figure 7: Dietary change recapitulates the effect of the NOD genetic background on a resistant mouse strain.
Figure 8: Molecular changes in the islets of patients with T2D mirror the processes altered in NOD mice.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Protein Data Bank

References

  1. Wicker, L.S. et al. Type 1 diabetes genes and pathways shared by humans and NOD mice. J. Autoimmun. 25 (suppl.) 29–33 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Islam, M.S. & Wilson, R.D. Experimentally induced rodent models of type 2 diabetes. Methods Mol. Biol. 933, 161–174 (2012).

    CAS  PubMed  Google Scholar 

  3. Butler, A.E. et al. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52, 102–110 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Bradfield, J.P. et al. A genome-wide meta-analysis of six type 1 diabetes cohorts identifies multiple associated loci. PLoS Genet. 7, e1002293 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) Consortium, Asian Genetic Epidemiology Network Type 2 Diabetes (AGEN-T2D) Consortium, South Asian Type 2 Diabetes (SAT2D) Consortium, Mexican American Type 2 Diabetes (MAT2D) Consortium & Type 2 Diabetes Genetic Exploration by Nex-generation sequencing in muylti-Ethnic Samples (T2D-GENES) Consortium. Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility. Nat. Genet. 46, 234–244 (2014).

  6. Fortune, M.D. et al. Statistical colocalization of genetic risk variants for related autoimmune diseases in the context of common controls. Nat. Genet. 47, 839–846 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Senée, V. et al. Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism. Nat. Genet. 38, 682–687 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Delépine, M. et al. EIF2AK3, encoding translation initiation factor 2-α kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat. Genet. 25, 406–409 (2000).

    Article  PubMed  Google Scholar 

  9. Janssens, S., Pulendran, B. & Lambrecht, B.N. Emerging functions of the unfolded protein response in immunity. Nat. Immunol. 15, 910–919 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tersey, S.A. et al. Islet beta-cell endoplasmic reticulum stress precedes the onset of type 1 diabetes in the nonobese diabetic mouse model. Diabetes 61, 818–827 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Engin, F. et al. Restoration of the unfolded protein response in pancreatic beta cells protects mice against type 1 diabetes. Sci. Transl. Med. 5, 211ra156 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Back, S.H. & Kaufman, R.J. Endoplasmic reticulum stress and type 2 diabetes. Annu. Rev. Biochem. 81, 767–793 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Planas, R., Pujol-Borrell, R. & Vives-Pi, M. Global gene expression changes in type 1 diabetes: insights into autoimmune response in the target organ and in the periphery. Immunol. Lett. 133, 55–61 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. Chaparro, R.J. et al. Nonobese diabetic mice express aspects of both type 1 and type 2 diabetes. Proc. Natl. Acad. Sci. USA 103, 12475–12480 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liston, A. et al. Generalized resistance to thymic deletion in the NOD mouse; a polygenic trait characterized by defective induction of Bim. Immunity 21, 817–830 (2004).

    CAS  PubMed  Google Scholar 

  16. Liston, A., Lesage, S., Wilson, J., Peltonen, L. & Goodnow, C.C. Aire regulates negative selection of organ-specific T cells. Nat. Immunol. 4, 350–354 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Hill, S.W., Kipp, D.E., Melchers, I., Frelinger, J.A. & Sercarz, E.E. Multiple H-2 and non-H-2 genes controlling the antilysozyme response: alternative gene constellations can lead to responsiveness. Eur. J. Immunol. 10, 384–391 (1980).

    Article  CAS  PubMed  Google Scholar 

  18. Izumi, T. et al. Dominant negative pathogenesis by mutant proinsulin in the Akita diabetic mouse. Diabetes 52, 409–416 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Iwawaki, T., Akai, R., Kohno, K. & Miura, M. A transgenic mouse model for monitoring endoplasmic reticulum stress. Nat. Med. 10, 98–102 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Wicker, L.S., Todd, J.A. & Peterson, L.B. Genetic control of autoimmune diabetes in the NOD mouse. Annu. Rev. Immunol. 13, 179–200 (1995).

    Article  CAS  PubMed  Google Scholar 

  21. Puebla-Osorio, N. et al. A novel Ku70 function in colorectal homeostasis separate from nonhomologous end joining. Oncogene 33, 2748–2757 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Tavana, O., Puebla-Osorio, N., Sang, M. & Zhu, C. Absence of p53-dependent apoptosis combined with nonhomologous end-joining deficiency leads to a severe diabetic phenotype in mice. Diabetes 59, 135–142 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Nijnik, A. et al. DNA repair is limiting for haematopoietic stem cells during ageing. Nature 447, 686–690 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Nogueira, T.C. et al. GLIS3, a susceptibility gene for type 1 and type 2 diabetes, modulates pancreatic beta cell apoptosis via regulation of a splice variant of the BH3-only protein Bim. PLoS Genet. 9, e1003532 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lindahl, M. et al. MANF is indispensable for the proliferation and survival of pancreatic beta cells. Cell Reports 7, 366–375 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Hall, E. et al. Effects of palmitate on genome-wide mRNA expression and DNA methylation patterns in human pancreatic islets. BMC Med. 12, 103 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sone, H. & Kagawa, Y. Pancreatic beta cell senescence contributes to the pathogenesis of type 2 diabetes in high-fat diet–induced diabetic mice. Diabetologia 48, 58–67 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Fadista, J. et al. Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism. Proc. Natl. Acad. Sci. USA 111, 13924–13929 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Leiter, E.H. et al. Unexpected functional consequences of xenogeneic transgene expression in beta-cells of NOD mice. Diabetes Obes. Metab. 9 (suppl. 2), 14–22 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. McCullough, K.D., Martindale, J.L., Klotz, L.O., Aw, T.Y. & Holbrook, N.J. Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol. Cell. Biol. 21, 1249–1259 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Apostolou, A., Shen, Y., Liang, Y., Luo, J. & Fang, S. Armet, a UPR-upregulated protein, inhibits cell proliferation and ER stress–induced cell death. Exp. Cell Res. 314, 2454–2467 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Davenport, E.L. et al. Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood 110, 2641–2649 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Neuber, O., Jarosch, E., Volkwein, C., Walter, J. & Sommer, T. Ubx2 links the Cdc48 complex to ER-associated protein degradation. Nat. Cell Biol. 7, 993–998 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Leitman, J. et al. Herp coordinates compartmentalization and recruitment of HRD1 and misfolded proteins for ERAD. Mol. Biol. Cell 25, 1050–1060 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Liston, A. et al. Gene dosage—limiting role of Aire in thymic expression, clonal deletion, and organ-specific autoimmunity. J. Exp. Med. 200, 1015–1026 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lesage, S. et al. Failure to censor forbidden clones of CD4 T cells in autoimmune diabetes. J. Exp. Med. 196, 1175–1188 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wick, G. et al. The role of genetically-determined primary alterations of the target organ in the development of spontaneous autoimmune thyroiditis in obese strain (OS) chickens. Immunol. Rev. 94, 113–136 (1986).

    Article  CAS  PubMed  Google Scholar 

  38. DIAMOND Project Group. Incidence and trends of childhood type 1 diabetes worldwide 1990–1999. Diabet. Med. 23, 857–866 (2006).

  39. Patterson, C.C., Dahlquist, G.G., Gyürüs, E., Green, A. & Soltész, G. Incidence trends for childhood type 1 diabetes in Europe during 1989–2003 and predicted new cases 2005–20: a multicentre prospective registration study. Lancet 373, 2027–2033 (2009).

    Article  PubMed  Google Scholar 

  40. TEDDY Study Group. The Environmental Determinants of Diabetes in the Young (TEDDY) Study. Ann. NY Acad. Sci. 1150, 1–13 (2008).

  41. Laybutt, D.R. et al. Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 50, 752–763 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Oprescu, A.I. et al. Free fatty acid–induced reduction in glucose-stimulated insulin secretion: evidence for a role of oxidative stress in vitro and in vivo. Diabetes 56, 2927–2937 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Betts, P., Mulligan, J., Ward, P., Smith, B. & Wilkin, T. Increasing body weight predicts the earlier onset of insulin-dependant diabetes in childhood: testing the 'accelerator hypothesis' (2). Diabet. Med. 22, 144–151 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Gale, E.A. & Gillespie, K.M. Diabetes and gender. Diabetologia 44, 3–15 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Donahue, R.P., Bean, J.A., Donahue, R.A., Goldberg, R.B. & Prineas, R.J. Insulin response in a triethnic population: effects of sex, ethnic origin, and body fat. Miami Community Health Study. Diabetes Care 20, 1670–1676 (1997).

    Article  CAS  PubMed  Google Scholar 

  46. Zhou, R., Tardivel, A., Thorens, B., Choi, I. & Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 11, 136–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Cervin, C. et al. Genetic similarities between latent autoimmune diabetes in adults, type 1 diabetes, and type 2 diabetes. Diabetes 57, 1433–1437 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Podolin, P.L. et al. I-E+ nonobese diabetic mice develop insulitis and diabetes. J. Exp. Med. 178, 793–803 (1993).

    Article  CAS  PubMed  Google Scholar 

  49. Akkaraju, S. et al. A range of CD4 T cell tolerance: partial inactivation to organ-specific antigen allows nondestructive thyroiditis or insulitis. Immunity 7, 255–271 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Mombaerts, P. et al. RAG-1–deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    Article  CAS  PubMed  Google Scholar 

  51. Ramsey, C. et al. Aire deficient mice develop multiple features of APECED phenotype and show altered immune response. Hum. Mol. Genet. 11, 397–409 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Prochazka, M., Gaskins, H.R., Shultz, L.D. & Leiter, E.H. The nonobese diabetic SCID mouse: model for spontaneous thymomagenesis associated with immunodeficiency. Proc. Natl. Acad. Sci. USA 89, 3290–3294 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lyons, P.A. et al. The NOD Idd9 genetic interval influences the pathogenicity of insulitis and contains molecular variants of Cd30, Tnfr2, and Cd137. Immunity 13, 107–115 (2000).

    Article  CAS  PubMed  Google Scholar 

  54. Lyons, P.A. et al. Congenic mapping of the type 1 diabetes locus, Idd3, to a 780-kb region of mouse chromosome 3: identification of a candidate segment of ancestral DNA by haplotype mapping. Genome Res. 10, 446–453 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hill, N.J. et al. NOD Idd5 locus controls insulitis and diabetes and overlaps the orthologous CTLA4/IDDM12 and NRAMP1 loci in humans. Diabetes 49, 1744–1747 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Mao, C., Dong, D., Little, E., Luo, S. & Lee, A.S. Transgenic mouse model for monitoring endoplasmic reticulum stress in vivo. Nat. Med. 10, 1013–1014 author reply 1014 (2004).

    Article  CAS  PubMed  Google Scholar 

  57. Kang, H.S. et al. Transcription factor Glis3, a novel critical player in the regulation of pancreatic beta-cell development and insulin gene expression. Mol. Cell. Biol. 29, 6366–6379 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Li, D.S., Yuan, Y.H., Tu, H.J., Liang, Q.L. & Dai, L.J. A protocol for islet isolation from mouse pancreas. Nat. Protoc. 4, 1649–1652 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Gysemans, C.A. et al. Disruption of the γ-interferon signaling pathway at the level of signal transducer and activator of transcription-1 prevents immune destruction of beta-cells. Diabetes 54, 2396–2403 (2005).

    Article  CAS  PubMed  Google Scholar 

  60. Kornete, M., Beauchemin, H., Polychronakos, C. & Piccirillo, C.A. Pancreatic islet cell phenotype and endocrine function throughout diabetes development in non-obese diabetic mice. Autoimmunity 46, 259–268 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Robinson, M.D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ghesquiere, B. et al. Redox proteomics of protein-bound methionine oxidation. Mol. Cell Proteomics 10, M110.006866 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Thimiri Govinda Raj, D.B. et al. A novel strategy for the comprehensive analysis of the biomolecular composition of isolated plasma membranes. Mol. Syst. Biol. 7, 541 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Helsens, K. et al. ms_lims, a simple yet powerful open source laboratory information management system for MS-driven proteomics. Proteomics 10, 1261–1264 (2010).

    Article  CAS  PubMed  Google Scholar 

  69. Sibanda, B.L. et al. Crystal structure of an Xrcc4–DNA ligase IV complex. Nat. Struct. Biol. 8, 1015–1019 (2001).

    Article  CAS  PubMed  Google Scholar 

  70. Jorgensen, W.L., Chandrasekhar, J., Madura, J.D., Impey, R.W. & Klein, M.L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 79, 926–935 (1983).

    Article  CAS  Google Scholar 

  71. Duan, Y. et al. A point-charge force field for molecular mechanics simulations of proteins based on condensed-phase quantum mechanical calculations. J. Comput. Chem. 24, 1999–2012 (2003).

    Article  CAS  PubMed  Google Scholar 

  72. Cheatham, T.E., Miller, J.L., Fox, T., Darden, T.A. & Kollman, P.A. Molecular-dynamics simulations on solvated biomolecular systems—the particle mesh Ewald method leads to stable trajectories of DNA, RNA, and proteins. J. Am. Chem. Soc. 117, 4193–4194 (1995).

    Article  CAS  Google Scholar 

  73. Ceriotti, M., Bussi, G. & Parrinello, M. Langevin equation with colored noise for constant-temperature molecular dynamics simulations. Phys. Rev. Lett. 102, 020601 (2009).

    Article  CAS  PubMed  Google Scholar 

  74. Feller, S.E., Zhang, Y.H., Pastor, R.W. & Brooks, B.R. Constant-pressure molecular-dynamics simulation—the Langevin piston method. J. Chem. Phys. 103, 4613–4621 (1995).

    Article  CAS  Google Scholar 

  75. Ryckaert, J.P., Ciccotti, G. & Berendsen, H.J.C. Numerical-integration of Cartesian equations of motion of a system with constraints—molecular-dynamics of N-alkanes. J. Comput. Phys. 23, 327–341 (1977).

    Article  CAS  Google Scholar 

  76. Miyamoto, S. & Kollman, P.A. Settle—an analytical version of the shake and rattle algorithm for rigid water models. J. Comput. Chem. 13, 952–962 (1992).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank P. Jeggo, C. Mathieu, D. Gray and A. Goris for critical insights and D. Pombal, J. Sreenivasan, A. Bullman, M. Koina and T. Dagpo for technical assistance. We acknowledge the Human Tissue Laboratory (HTL) of the Lund University Diabetes Centre (LUDC) for providing high-quality data from human pancreatic islets. This work was supported by the VIB, European Research Council (ERC) and a Juvenile Diabetes Research Foundation (JDRF) Career Development Award (A.L.) and by the National Health and Medical Research Council of Australia (project grant 1028108; C.J.N.). N.O. and V. Lyssenko acknowledge support by a Strategic Research Grant from the Swedish Research Council (2009-1039).

Author information

Authors and Affiliations

Authors

Contributions

A.L., S.M.S., S.L., N.P., C.J.N., D.R.L. and C.C.G. designed the study. J.D., L.T., S.S., V.D.-A., J.E.G.-P., E.P., D.D.M., D.F., L.O., J.V., G.C.-R., J.D., L.S., S.M.S., J.E.D. and A.L. performed the experiments. E.J.C., N.O., V. Lyssenko, V. Lagou, J.A., K.G., D.L., M.A.L. and A.L. analyzed results. A.M.J. provided reagents. A.L. wrote the manuscript. All authors discussed results and read and approved the manuscript.

Corresponding author

Correspondence to Adrian Liston.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Equivalent islet insHEL expression on the B10 and NOD backgrounds.

RNA-seq was performed on the islets of wild-type (wt) B10k.Rag−/−, B10k.Rag−/−.insHEL, wt NODk.Rag−/− and (pre-diabetic) NODk.Rag−/−.insHEL mice (n = 3/group). (a) Reads were aligned to a custom genome build combining chicken chromosome 1 with the mouse genome, with normalized counts displayed for chicken Lyz, the gene encoding HEL. (b) Expression of H2k1, of which the transmembrane region is included in the insHEL construct to encode a membrane anchor. Means ± s.e.m.

Supplementary Figure 2 No effect of insHEL insertion on chemokine gene expression from the endogenous locus.

RNA-seq was performed on the islets of B10k.Rag−/−, B10k.Rag−/−.insHEL, NODk.Rag−/− and (pre-diabetic) NODk.Rag−/−.insHEL mice (n = 3/group). Expression of chemokines is given for each strain. Means ± s.e.m.: *P < 0.05.

Supplementary Figure 3 No effect of insHEL insertion on gene expression from the endogenous locus.

The gene insertion site of insHEL was defined by the congenic region remaining in the backcross to the NOD background. The outer boundaries of the integration site were identified on chromosome 12 (64,900,000–71,900,000). RNA-seq was performed on the islets of B10k.Rag−/−, B10k.Rag−/−.insHEL, NODk.Rag−/− and (pre-diabetic) NODk.Rag−/−.insHEL mice (n = 3/group). (a) Expression of genes within the integration boundaries in B10k.Rag−/− islets. (b) For all genes with reliable expression detection within the boundaries, the effect of insHEL expression was assessed as the expression in transgenic islets as a percentage of the expression in non-transgenic islets on the B10k strain (black), NODk strain (white) or average of the two strains (gray). No genes in the interval showed a significant change in expression.

Supplementary Figure 4 Altered insulin expression in the islets of insHEL transgenic mice.

(a) Immunofluorescence analysis was performed on the islets of wild-type (wt) B10k.Rag−/−, B10k.Rag−/−.insHEL, wt NODk.Rag−/− and (pre-diabetic) NODk.Rag−/−.insHEL mice with a polyclonal antibody to insulin, antibody to glucagon and DAPI. Staining is representative of three experiments. Scale bar, 100 μm. (b,c) RNA-seq analysis was performed on the islets of B10k.Rag−/−, B10k.Rag−/−.insHEL, NODk.Rag−/− and (pre-diabetic) NODk.Rag−/−.insHEL mice (n = 3/group). Expression is shown for Ins1 (b) and Ins2 (c). (d,e) ERAI mice were crossed to B10k.Rag−/−.insHEL and NODk.Rag−/−.insHEL mice, and islets were analyzed by flow cytometry. Histograms are shown of insulin (d) and side scatter in insulin-expressing beta cells (e). Results are representative of three experiments. Means ± s.e.m. **p<0.001.

Supplementary Figure 5 Transgene-induced beta cell stress results in disturbed insulin processing.

(a,b) Islets from B10k.Rag0/0, B10k.Rag0/0.insHEL, NODk.Rag0/0 and NODk.Rag0/0.insHEL mice were cultured in vitro in the presence of a high (25 mM) glucose concentration (n = 5/group) and assessed for insulin (a) and proinsulin (b) secretion by ELISA. (c,d) RNA-seq analysis was performed on the islets of B10k.Rag0/0, B10k.Rag0/0.insHEL, NODk.Rag0/0 and (pre-diabetic) NODk.Rag0/0.insHEL mice (n = 3/group). Expression is shown for Pcsk1 (prohormone convertase 1-3) (c) and Pcsk2 (prohormone convertase 2) (d). (e,f) Pancreas immunofluorescence with a polyclonal antibody to insulin, antibody to PC1 and PC3, and DAPI, with mice of the B10k.Rag0/0 and NODk.Rag0/0 backgrounds. Quantification is shown for islet raw fluorescence in the PC1-PC3 channel (n = 10, 16, 17, 16) (e), with representative sections (f). Scale bar, 100 μm. (gi) Quantification of immunoblot analysis of islets from B10k, B10k.insHEL, NODk and NODk.insHEL mice for PC1-PC3 (g) and PC2 (h), with representative blots (n = 3/group) (i). Means ± s.e.m.: *P < 0.05, **P < 0.001, ***P < 0.0001.

Supplementary Figure 6 Glucose intolerance in insHEL transgenic mice.

(a,b) Blood glucose levels following a glucose tolerance test in B10k.Rag−/− (n = 15) and B10k.Rag−/−.insHEL (n = 17) mice at 12 (a) and 24 (b) weeks of age. (c,d) Blood glucose levels following a glucose tolerance test in (B10k × NODk).F1.Rag−/− (n = 11) and pre-diabetic (B10k × NODk)F1.Rag−/−.insHEL (n = 19) mice at 12 (c) and 24 (d) weeks of age. (e) Blood insulin levels following a glucose tolerance test in B10k (n = 5), B10k.insHEL (n = 4), NODk (n = 15) and NODk.insHEL (n = 9) mice at 12 weeks of age. Means ± s.e.m.: *P < 0.05, **P < 0.001, ***P < 0.0001.

Supplementary Figure 7 Sex hormones control susceptibility to islet stress and diabetes.

(a) Table of the incidence of diabetes at 26 weeks of age in female insHEL transgenic mice. (b) Representative electron microscopy images of beta cells from male and female NODk and NODk.insHEL mice at 12 weeks of age. Scale bar, 5 μm. (c) Images were used to assess the number of insulin granules per cellular cross-section (n = 3/group). (d,e) ERAI mice were crossed to NODk.Rag−/−.insHEL mice, and islets were analyzed by flow cytometry. Histograms show side scatter in insulin-expressing beta cells (d) and insulin (e). Results are representative of three experiments. (fh) Fasting serum samples from male and female NODk and NODk.insHEL mice at 24 weeks of age were assessed by ELISA for insulin (n = 11, 11, 12, 7) (f), proinsulin (n = 11, 11, 12, 8) (g) and C-peptide (n = 11, 11, 9, 8) (h). (i,j) Pancreas immunofluorescence with a polyclonal antibody to insulin, antibody to PC1 and PC3, and DAPI, with female NODk.Rag−/− mice. Representative sections (scale bar, 100 μm) are shown (i), with quantification of islet raw fluorescence in the PC1-PC3 channel (n = 15, 25) (j). (k) Blood glucose levels in 12-week-old female NODk (n = 10) and NODk.insHEL (n = 9) mice following glucose tolerance test. (l) Average number of islets per pancreatic section in female NODk and NODk.insHEL mice at 28 weeks of age (n = 3 mice/group). (m) Incidence of diabetes in NODk insHEL transgenic male mice, with (n = 24) and without (n = 24) castration. Means ± s.e.m.: *P < 0.05, **P < 0.001, ***P < 0.0001.

Supplementary Figure 8 Expression of insHEL activates similar gene sets in B10 and NOD islets.

(a) Genes with significant expression changes between B10k.Rag−/− and NODk.Rag−/− islets were plotted for average expression. Outliers are annotated, Scg2 is not shown. (b,c) Cytoscape enrichment map for significant gene sets (FDR < 0.001) between B10k.Rag−/− islets and B10k.Rag−/−.insHEL islets (b) or NODk.Rag−/− islets and NODk.Rag−/−.insHEL islets (c). UPR and Xbp1 response gene sets are shown in yellow.

Supplementary Figure 9 Expression of insHEL induces similar transcriptional changes in qualitatively different B10 and NOD islets.

(a,b) Expression of genes significantly upregulated between B10k.Rag−/− islets and B10k.Rag−/−.insHEL islets (a) or between NODk.Rag−/− islets and NODk.Rag−/−.insHEL islets (b), with the average expression shown across all samples. Data represented are normalized to the expression in B10k.Rag−/− islets, with the average percentage and outliers annotated. H2k1 is not shown. (c,d) Expression of genes significantly downregulated between B10k.Rag−/− islets and B10k.Rag−/−.insHEL islets (c) or between NODk.Rag−/− islets and NODk.Rag−/−.insHEL islets (d), with the average expression shown across all samples. Data represented are normalized to the expression in B10k.Rag−/− islets, with the average percentage and outliers annotated.

Supplementary Figure 10 Molecular modeling of the effect of p.Ala27Thr and p.Glu125Asp mutations on Xrcc4 stability.

Simulation of the movements of the DNA ligase IV complex during 100-ns simulation. (a,b) Representative snapshots of the B10 allele (a) and NOD allele (b) for the DNA ligase IV complex extracted from the two trajectories at 0 ns, 60 ns, 80 ns and 100 ns. (c) Root-mean-square deviation of the BRCT2 domain of ligase 4 calculated with respect to the fixed position of BRCT1. Movements after the initial 70 ns of equilibrium were considered reliable. The black and gray lines represent ligase IV from the B10 and NOD alleles, respectively. Results are representative of four simulations.

Supplementary Figure 11 DNA ligase 4 hypomorph enhances susceptibility to diabetes.

B10k.insHEL mice were intercrossed with the ligase 4 hypomorph Tyr288Cys, and diabetes incidence was analyzed in B10 (n = 6), B10.Lig4Y228C/+ (n = 5), B10.insHEL (n = 7) and B10.Lig4Y228C/+.insHEL (n = 11) littermates. **P < 0.01.

Supplementary Figure 12 Transgene-induced beta cell stress results in disturbed insulin processing and glucose intolerance on the Glis3 heterozygous background.

(ad) Fasting serum samples from wild-type (wt) B10 (n = 30), wt B10.Glis3+/– (n = 6), B10.insHEL (n = 51) and B10.Glis3+/–.insHEL (n = 7) mice at 10 weeks of age were assessed by ELISA for insulin (a), proinsulin (b), C-peptide (c) and proinsulin/insulin ratio (d). (e,f) Blood glucose levels in 12-week-old B10 (n = 6), B10.insHEL (n = 1), B10.Glis3+/– (n = 3) and (non-diabetic) B10.Glis3+/–.insHEL (n = 4) mice following glucose tolerance test (n = 28, 47, 9, 21) (e) or insulin tolerance test (n = 8, 17, 3, 13) (f). Means ± s.e.m.: *P < 0.05, **P < 0.001, ***P < 0.0001.

Supplementary Figure 13 Expression of GLIS3 and MANF in the islets of patients with T2D.

mRNA expression in human pancreatic islets from healthy individuals (n = 105) and individuals diagnosed with T2D (n = 14) was assessed through RNA-seq analysis. (a,b) Expression of GLIS3 (a) and MANF (b) in islets from healthy individuals as compared to islets from individuals with T2D. The median and interquartile range (box) are shown, with error bars indicating 1.5xIQR (interquartile range). Individual values are shown if beyond 1.5xIQR.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13 and Supplementary Tables 1 and 2. (PDF 4390 kb)

Supplementary Data Set 1

Transcriptional analysis of the effect of insHEL on the B10 and NOD backgrounds. (XLSX 17127 kb)

Supplementary Data Set 2

Network analysis of the effect of insHEL on the B10 and NOD backgrounds. (XLSX 2954 kb)

Supplementary Data Set 3

Proteomics analysis of the effect of insHEL on the B10 and NOD backgrounds. (XLSX 153 kb)

Supplementary Data Set 4

Genomic variation analysis of islet-expressed genes on the B10 and NOD backgrounds. (XLSX 1842 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dooley, J., Tian, L., Schonefeldt, S. et al. Genetic predisposition for beta cell fragility underlies type 1 and type 2 diabetes. Nat Genet 48, 519–527 (2016). https://doi.org/10.1038/ng.3531

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3531

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