Skip to main content

Advertisement

Log in

β-cell failure as a complication of diabetes

  • Published:
Reviews in Endocrine and Metabolic Disorders Aims and scope Submit manuscript

Abstract

Type 2 diabetes mellitus is a complex disease characterized by β-cell failure in the setting of insulin resistance. In early stages of the disease, pancreatic β-cells adapt to insulin resistance by increasing mass and function. As nutrient excess persists, hyperglycemia and elevated free fatty acids negatively impact β-cell function. This happens by numerous mechanisms, including the generation of reactive oxygen species, alterations in metabolic pathways, increases in intracellular calcium and the activation of endoplasmic reticulum stress. These processes adversely affect β-cells by impairing insulin secretion, decreasing insulin gene expression and ultimately causing apoptosis. In this review, we will first discuss the regulation of β-cell mass during normal conditions. Then, we will discuss the mechanisms of β-cell failure, including glucotoxicity, lipotoxicity and endoplasmic reticulum stress. Further research into mechanisms will reveal the key modulators of β-cell failure and thus identify possible novel therapeutic targets. Type 2 diabetes mellitus is a multifactorial disease that has greatly risen in prevalence in part due to the obesity and inactivity that characterize the modern Western lifestyle. Pancreatic β-cells possess the potential to greatly expand their function and mass in both physiologic and pathologic states of nutrient excess and increased insulin demand. β-cell response to nutrient excess occurs by several mechanisms, including hypertrophy and proliferation of existing β-cells, increased insulin production and secretion, and formation of new β-cells from progenitor cells [1, 2]. Failure of pancreatic β-cells to adequately expand in settings of increased insulin demand results in hyperglycemia and diabetes. In this review, we will first discuss the factors involved in β-cell growth and then discuss the mechanisms by which β-cell expansion fails and leads to β-cell failure and diabetes (Fig. 1).

The mechanisms by which -cell failure and apoptosis occur are complex, not completely unraveled and involve the interplay of numerous factors and conditions. These factors are summarized in this figure. Glucotoxicity and lipotoxicity lead to the production of ROS, which activate JNK. JNK activity leads to a decrease in IRS signaling and may directly be involved in decreased Pdx-1 activity by translocation from the nucleus to the cytoplasm [183]. In addition, glucose and FA have both been found to induce ER stress. Chronic glucose elevation inhibits FA oxidation and favors the generation of ceramide and lipid partitioning, which ultimately results in β-cell dysfunction and apoptosis. AMPK activation promotes fatty acid oxidation by phosphorylation and inhibition of acetyl-CoA carboxylase or via down regulation of the transcription factor sterol-regulatory-element-binding-protein-1c (SREBP1c) and subsequent decreases in acetyl-CoA carboxylase. Glucose and FA also activate the UPR response and induce ER stress. The ER stress response and its effectors are activated in response to misfolded proteins in order to protect β-cells from apoptosis; however, activation of these processes under conditions of long-term elevation of FFA and glucose can lead to β-cell dysfunction and ultimately apoptosis. Activation of ER stress leads to inhibition of insulin mRNA and protein expression and may also be pro-apoptotic. The mechanisms for induction of apoptosis by ER stress are not completely known, but induction of CHOP is an important component. In addition, induction of ATF3 and SREBP can downregulate IRS signaling by repressing IRS2 transcription. One interesting finding is that inhibition of IRS signaling seems to be a common pathway induced by the majority of the mechanisms described for β-cell failure. An additional event is the increase in mTOR signaling by nutrient excess (glucose). This results in negative feedback inhibition on IRS1 and possibly IRS2 by activation of S6K signaling. The decrease in IRS signaling induces GSK3β and Foxo1 function. Activation of these molecules ultimately reduces Pdx1 levels and increases the levels of the cell cycle inhibitor p27

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Bonner-Weir S, Deery D, Leahy JL, Weir GC. Compensatory growth of pancreatic beta-cells in adult rats after short-term glucose infusion. Diabetes 1989;38:49–53.

    Article  PubMed  CAS  Google Scholar 

  2. Bruning JC, Winnay J, Bonner-Weir S, Taylor SI, Accili D, Kahn CR. Development of a novel polygenic model of NIDDM in mice heterozygous for IR and IRS-1 null alleles. Cell 1997;88:561–72.

    Article  PubMed  CAS  Google Scholar 

  3. Parsons JA, Brelje TC, Sorenson RL. Adaptation of islets of Langerhans to pregnancy: increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology 1992;130:1459–66.

    Article  PubMed  CAS  Google Scholar 

  4. Scaglia L, Smith FE, Bonner-Weir S. Apoptosis contributes to the involution of beta cell mass in the post partum rat pancreas. Endocrinology 1995;136:5461–8.

    Article  PubMed  CAS  Google Scholar 

  5. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren J-M, Previs S, et al. Disruption of IRS-2 causes type 2 diabetes in mice. Nature 1998;391:900–3.

    Article  PubMed  CAS  Google Scholar 

  6. Kido Y, Burks DJ, Withers D, Bruning JC, Kahn CR, White MF, et al. Tissue-specific insulin resistance in mice with mutations in the insulin receptor, IRS-1, and IRS-2. J Clin Invest 2000;105:199–205.

    Article  PubMed  CAS  Google Scholar 

  7. Xu G, Stoffers DA, Habener JF, Bonner-Weir S. Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats. Diabetes 1999;48:2270–6.

    Article  PubMed  CAS  Google Scholar 

  8. Bonner-Weir S, Baxter LA, Schuppin GT, Smith FE. A second pathway for regeneration of adult exocrine and endocrine pancreas. A possible recapitulation of embryonic development. Diabetes 1993;42:1715–20.

    Article  PubMed  CAS  Google Scholar 

  9. Dor Y, Brown J, Martinez OI, Melton DA. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 2004;429:41–6.

    Article  PubMed  CAS  Google Scholar 

  10. Bonner-Weir S, Inada A, Yatoh S, Li WC, Aye T, Toschi E, Sharma A. Transdifferentiation of pancreatic ductal cells to endocrine beta-cells. Biochem Soc Trans 2008;36:353–6.

    Article  PubMed  CAS  Google Scholar 

  11. Cozar-Castellano I, Fiaschi-Taesch N, Bigatel TA, Takane KK, Garcia-Ocana A, Vasavada R, et al. Molecular Control of Cell Cycle Progression in the Pancreatic {beta}-Cell. Endocr Rev 2006;27:356–70.

    Article  PubMed  CAS  Google Scholar 

  12. Zhang X, Gaspard JP, Mizukami Y, Li J, Graeme-Cook F, Chung DC. Overexpression of cyclin D1 in pancreatic beta-cells in vivo results in islet hyperplasia without hypoglycemia. Diabetes 2005;54:712–9.

    Article  PubMed  CAS  Google Scholar 

  13. Georgia S, Bhushan A. Beta cell replication is the primary mechanism for maintaining postnatal beta cell mass. J Clin Invest 2004;114:963–8.

    PubMed  CAS  Google Scholar 

  14. Kushner JA, Ciemerych MA, Sicinska E, Wartschow LM, Teta M, Long SY, et al. Cyclins D2 and D1 Are Essential for Postnatal Pancreatic {beta}-Cell Growth. Mol Cell Biol 2005;25:3752–62.

    Article  PubMed  CAS  Google Scholar 

  15. Rane SG, Dubus P, Mettus RV, Galbreath EJ, Boden G, Reddy EP, et al. Loss of Cdk4 expression causes insulin-deficient diabetes and Cdk4 activation results in beta-islet cell hyperplasia. Nat Genet 1999;22:44–52.

    Article  PubMed  CAS  Google Scholar 

  16. Cozar-Castellano I, Weinstock M, Haught M, Velazquez-Garcia S, Sipula D, Stewart AF. Evaluation of beta-cell replication in mice transgenic for hepatocyte growth factor and placental lactogen: comprehensive characterization of the G1/S regulatory proteins reveals unique involvement of p21cip. Diabetes 2006;55:70–7.

    Article  PubMed  CAS  Google Scholar 

  17. Serrano M, Hannon GJ, Beach D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature 1993;366:704–7.

    Article  PubMed  CAS  Google Scholar 

  18. Marzo N, Mora C, Fabregat ME, Martin J, Usac EF, Franco C, et al. Pancreatic islets from cyclin-dependent kinase 4/R24C (Cdk4) knockin mice have significantly increased beta cell mass and are physiologically functional, indicating that Cdk4 is a potential target for pancreatic beta cell mass regeneration in Type 1 diabetes. Diabetologia 2004;47(4):686–94.

    Article  PubMed  CAS  Google Scholar 

  19. Rachdi L, Balcazar N, Elghazi L, Barker DJ, Krits I, Kiyokawa H, et al. Differential effects of p27 in regulation of beta-cell mass during development, neonatal period, and adult life. Diabetes 2006;55:3520–8.

    Article  PubMed  CAS  Google Scholar 

  20. Uchida T, Nakamura T, Hashimoto N, Matsuda T, Kotani K, Sakaue H, et al. Deletion of Cdkn1b ameliorates hyperglycemia by maintaining compensatory hyperinsulinemia in diabetic mice. Nat Med 2005;11:175–82.

    Article  PubMed  CAS  Google Scholar 

  21. Tsutsui T, Hesabi B, Moons DS, Pandolfi PP, Hansel KS, Koff A, et al. Targeted disruption of CDK4 delays cell cycle entry with enhanced p27(Kip1) activity. Mol Cell Biol 1999;19:7011–9.

    PubMed  CAS  Google Scholar 

  22. Bonner-Weir S, Taneja M, Weir GC, Tatarkiewicz K, Song KH, Sharma A, et al. In vitro cultivation of human islets from expanded ductal tissue. Proc Natl Acad Sci U S A 2000;97:7999–8004.

    Article  PubMed  CAS  Google Scholar 

  23. Ramiya VK, Maraist M, Arfors KE, Schatz DA, Peck AB, Cornelius JG. Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells. Nat Med 2000;6:278–82.

    Article  PubMed  CAS  Google Scholar 

  24. Rooman I, Lardon J, Bouwens L. Gastrin stimulates beta-cell neogenesis and increases islet mass from transdifferentiated but not from normal exocrine pancreas tissue. Diabetes 2002;51:686–90.

    Article  PubMed  CAS  Google Scholar 

  25. Li L, Seno M, Yamada H, Kojima I. Betacellulin improves glucose metabolism by promoting conversion of intraislet precursor cells to beta-cells in streptozotocin-treated mice. Am J Physiol Endocrinol Metab 2003;285:E577–83.

    PubMed  CAS  Google Scholar 

  26. Cnop M, Welsh N, Jonas JC, Jorns A, Lenzen S, Eizirik DL. Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 2005;54(Suppl 2):S97–107.

    Article  PubMed  CAS  Google Scholar 

  27. Kloppel G, Lohr M, Habich K, Oberholzer M, Heitz PU. Islet pathology and the pathogenesis of type 1 and type 2 diabetes mellitus revisited. Surv Synth Pathol Res 1985;4:110–25.

    Article  PubMed  CAS  Google Scholar 

  28. Clark A, Wells CA, Buley ID, Cruickshank JK, Vanhegan RI, Matthews DR, et al. Islet amyloid, increased A-cells, reduced B-cells and exocrine fibrosis: quantitative changes in the pancreas in type 2 diabetes. Diabetes Res 1988;9:151–9.

    PubMed  CAS  Google Scholar 

  29. Wajchenberg BL. Beta-cell failure in diabetes and preservation by clinical treatment. Endocr Rev 2007;28:187–218.

    Article  PubMed  CAS  Google Scholar 

  30. Donath MY, Halban PA. Decreased beta-cell mass in diabetes: significance, mechanisms and therapeutic implications. Diabetologia 2004;47:581–9.

    Article  PubMed  CAS  Google Scholar 

  31. Swenne I. The role of glucose in the in vitro regulation of cell cycle kinetics and proliferation of fetal pancreatic B-cells. Diabetes 1982;31:754–60.

    PubMed  CAS  Google Scholar 

  32. Chick WL. Beta cell replication in rat pancreatic monolayer cultures. Effects of glucose, tolbutamide, glucocorticoid, growth hormone and glucagon. Diabetes 1973;22:687–93.

    PubMed  CAS  Google Scholar 

  33. Paris M, Bernard-Kargar C, Berthault MF, Bouwens L, Ktorza A. Specific and combined effects of insulin and glucose on functional pancreatic beta-cell mass in vivo in adult rats. Endocrinology 2003;144:2717–27.

    Article  PubMed  CAS  Google Scholar 

  34. Bernard C, Berthault MF, Saulnier C, Ktorza A. Neogenesis vs. apoptosis As main components of pancreatic beta cell ass changes in glucose-infused normal and mildly diabetic adult rats. FasebJ 1999;13:1195–205.

    CAS  Google Scholar 

  35. Alonso LC, Yokoe T, Zhang P, Scott DK, Kim SK, O’Donnell CP, et al. Glucose infusion in mice: a new model to induce beta-cell replication. Diabetes 2007;56:1792–801.

    Article  PubMed  CAS  Google Scholar 

  36. Hugl SR, White MF, Rhodes CJ. Insulin-like growth factor I (IGF-I)-stimulated pancreatic beta-cell growth is glucose-dependent. Synergistic activation of insulin receptor substrate-mediated signal transduction pathways by glucose and IGF-I in INS-1 cells. J Biol Chem 1998;273:17771–9.

    Article  PubMed  CAS  Google Scholar 

  37. Kulkarni RN, Bruning JC, Winnay JN, Postic C, Magnuson MA, Kahn CR. Tissue-specific knockout of the insulin receptor in pancreatic beta cells creates an insulin secretory defect similar to that in type 2 diabetes. Cell 1999;96:329–39.

    Article  PubMed  CAS  Google Scholar 

  38. Ohsugi M, Cras-Meneur C, Zhou Y, Warren W, Bernal-Mizrachi E, Permutt MA. Glucose and insulin treatment of insulinoma cells results in transcriptional regulation of a common set of genes. Diabetes 2004;53:1496–508.

    Article  PubMed  CAS  Google Scholar 

  39. Tuttle RL, Gill NS, Pugh W, Lee JP, Koeberlein B, Furth EE, et al. Regulation of pancreatic beta-cell growth and survival by the serine/threonine protein kinase Akt1/PKBalpha. Nat Med 2001;7:1133–7.

    Article  PubMed  CAS  Google Scholar 

  40. Bernal-Mizrachi E, Wen W, Stahlhut S, Welling CM, Permutt MA. Islet beta cell expression of constitutively active Akt1/PKB alpha induces striking hypertrophy, hyperplasia, and hyperinsulinemia. J Clin Invest 2001;108:1631–8.

    PubMed  CAS  Google Scholar 

  41. Heit JJ, Apelqvist AA, Gu X, Winslow MM, Neilson JR, Crabtree GR, et al. Calcineurin/NFAT signalling regulates pancreatic beta-cell growth and function. Nature 2006;443:345–9.

    Article  PubMed  CAS  Google Scholar 

  42. Jhala US, Canettieri G, Screaton RA, Kulkarni RN, Krajewski S, Reed J, et al. cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev 2003;17:1575–80.

    Article  PubMed  CAS  Google Scholar 

  43. Bernal-Mizrachi E, Wice B, Inoue H, Permutt MA. Activation of serum response factor in the depolarization induction of Egr-1 transcription in pancreatic islet beta-cells. J Biol Chem 2000;275:25681–9.

    Article  PubMed  CAS  Google Scholar 

  44. Sorenson RL, Stout LE. Prolactin receptors and JAK2 in islets of Langerhans: an immunohistochemical analysis. Endocrinology 1995;136:4092–8.

    Article  PubMed  CAS  Google Scholar 

  45. Brelje TC, Sorenson RL. Role of prolactin versus growth hormone on islet B-cell proliferation in vitro: implications for pregnancy. Endocrinology 1991;128:45–57.

    PubMed  CAS  Google Scholar 

  46. Brelje TC, Scharp DW, Lacy PE, Ogren L, Talamantes F, Robertson M, et al. Effect of homologous placental lactogens, prolactins, and growth hormones on islet B-cell division and insulin secretion in rat, mouse, and human islets: implication for placental lactogen regulation of islet function during pregnancy. Endocrinology 1993;132:879–87.

    Article  PubMed  CAS  Google Scholar 

  47. Vasavada RC, Garcia-Ocana A, Zawalich WS, Sorenson RL, Dann P, Syed M, et al. Targeted expression of placental lactogen in the beta cells of transgenic mice results in beta cell proliferation, islet mass augmentation, and hypoglycemia. J Biol Chem 2000;275:15399–406.

    Article  PubMed  CAS  Google Scholar 

  48. Freemark M, Avril I, Fleenor D, Driscoll P, Petro A, Opara E, et al. Targeted deletion of the PRL receptor: effects on islet development, insulin production, and glucose tolerance. Endocrinology 2002;143:1378–85.

    Article  PubMed  CAS  Google Scholar 

  49. Liu JL, Coschigano KT, Robertson K, Lipsett M, Guo Y, Kopchick JJ, et al. Disruption of growth hormone receptor gene causes diminished pancreatic islet size and increased insulin sensitivity in mice. Am J Physiol Endocrinol Metab 2004;287:E405–13.

    Article  PubMed  CAS  Google Scholar 

  50. Villanueva-Penacarrillo ML, Cancelas J, de Miguel F, Redondo A, Valin A, Valverde I, et al. Parathyroid hormone-related peptide stimulates DNA synthesis and insulin secretion in pancreatic islets. J Endocrinol 1999;163:403–8.

    Article  PubMed  CAS  Google Scholar 

  51. Fujinaka Y, Sipula D, Garcia-Ocana A, Vasavada RC. Characterization of mice doubly transgenic for parathyroid hormone-related protein and murine placental lactogen: a novel role for placental lactogen in pancreatic beta-cell survival. Diabetes 2004;53:3120–30.

    Article  PubMed  CAS  Google Scholar 

  52. Vasavada RC, Gonzalez-Pertusa JA, Fujinaka Y, Fiaschi-Taesch N, Cozar-Castellano I, Garcia-Ocana A. Growth factors and beta cell replication. Int J Biochem Cell Biol. 2005;

  53. Matsumoto K, Nakamura T. Emerging multipotent aspects of hepatocyte growth factor. J Biochem 1996;119:591–600.

    PubMed  CAS  Google Scholar 

  54. Garcia-Ocana A, Vasavada RC, Cebrian A, Reddy V, Takane KK, Lopez-Talavera JC, et al. Transgenic overexpression of hepatocyte growth factor in the beta-cell markedly improves islet function and islet transplant outcomes in mice. Diabetes 2001;50:2752–62.

    Article  PubMed  CAS  Google Scholar 

  55. Furge KA, Zhang YW, Vande Woude GF. Met receptor tyrosine kinase: enhanced signaling through adapter proteins. Oncogene 2000;19:5582–9.

    Article  PubMed  CAS  Google Scholar 

  56. Calvo EL, Boucher C, Pelletier G, Morisset J. Ontogeny of hepatocyte growth factor and c-met/hgf receptor in rat pancreas. Biochem Biophys Res Commun 1996;229:257–63.

    Article  PubMed  CAS  Google Scholar 

  57. Dai C, Li Y, Yang J, Liu Y. Hepatocyte growth factor preserves beta cell mass and mitigates hyperglycemia in streptozotocin-induced diabetic mice. J Biol Chem 2003;278:27080–7.

    Article  PubMed  CAS  Google Scholar 

  58. Roccisana J, Reddy V, Vasavada RC, Gonzalez-Pertusa JA, Magnuson MA, Garcia-Ocana A. Targeted inactivation of hepatocyte growth factor receptor c-met in beta-cells leads to defective insulin secretion and GLUT-2 downregulation without alteration of beta-cell mass. Diabetes 2005;54:2090–102.

    Article  PubMed  CAS  Google Scholar 

  59. Vasavada RC, Gonzalez-Pertusa JA, Fujinaka Y, Fiaschi-Taesch N, Cozar-Castellano I, Garcia-Ocana A. Growth factors and beta cell replication. Int J Biochem Cell Biol 2006;38:931–50.

    Article  PubMed  CAS  Google Scholar 

  60. Nielsen JHGE, Møldrup A, Friedrichsen BN, Billestrup N, Hansen JA, Lee YC, et al. Regulation of beta-cell mass by hormones and growth factors. Diabetes 2001;50:S25–9.

    Article  PubMed  CAS  Google Scholar 

  61. Friedrichsen BN, Richter HE, Hansen JA, Rhodes CJ, Nielsen JH, Billestrup N, et al. Signal transducer and activator of transcription 5 activation is sufficient to drive transcriptional induction of cyclin D2 gene and proliferation of rat pancreatic beta-cells. Mol Endocrinol 2003;17:945–58.

    Article  PubMed  CAS  Google Scholar 

  62. Zhang B, Hosaka M, Sawada Y, Torii S, Mizutani S, Ogata M, et al. Parathyroid hormone-related protein induces insulin expression through activation of MAP kinase-specific phosphatase-1 that dephosphorylates c-Jun NH2-terminal kinase in pancreatic beta-cells. Diabetes 2003;52:2720–30.

    Article  PubMed  CAS  Google Scholar 

  63. Vasavada RC, Wang L, Fujinaka Y, Takane KK, Rosa TC, Mellado-Gil JM, et al. Protein kinase C-zeta activation markedly enhances beta-cell proliferation: an essential role in growth factor mediated beta-cell mitogenesis. Diabetes 2007;56:2732–43.

    Article  PubMed  CAS  Google Scholar 

  64. Hogg J, Han VK, Clemmons DR, Hill DJ. Interactions of nutrients, insulin-like growth factors (IGFs) and IGF-binding proteins in the regulation of DNA synthesis by isolated fetal rat islets of Langerhans. J Endocrinol 1993;138:401–12.

    PubMed  CAS  Google Scholar 

  65. Sieradzki J, Fleck H, Chatterjee AK, Schatz H. Stimulatory effect of insulin-like growth factor-I on [3H]thymidine incorporation, DNA content and insulin biosynthesis and secretion of isolated pancreatic rat islets. J Endocrinol 1988;117:59–62.

    PubMed  CAS  Google Scholar 

  66. Swenne I, Hill DJ, Strain AJ, Milner RD. Growth hormone regulation of somatomedin C/insulin-like growth factor I production and DNA replication in fetal rat islets in tissue culture. Diabetes 1987;36:288–94.

    Article  PubMed  CAS  Google Scholar 

  67. George M, Ayuso E, Casellas A, Costa C, Devedjian JC, Bosch F. Beta cell expression of IGF-I leads to recovery from type 1 diabetes. J Clin Invest 2002;109:1153–63.

    PubMed  CAS  Google Scholar 

  68. Petrik J, Pell JM, Arany E, McDonald TJ, Dean WL, Reik W, et al. Overexpression of insulin-like growth factor-II in transgenic mice is associated with pancreatic islet cell hyperplasia. Endocrinology 1999;140:2353–63.

    Article  PubMed  CAS  Google Scholar 

  69. Ohsugi M, Cras-Meneur C, Zhou Y, Bernal-Mizrachi E, Johnson JD, Luciani DS, et al. Reduced expression of the insulin receptor in mouse insulinoma (MIN6) cells reveals multiple roles of insulin signaling in gene expression, proliferation, insulin content, and secretion. J Biol Chem 2005;280:4992–5003.

    Article  PubMed  CAS  Google Scholar 

  70. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, et al. Disruption of IRS-2 causes type 2 diabetes in mice. Nature 1998;391:900–4.

    Article  PubMed  CAS  Google Scholar 

  71. Kulkarni RN. New insights into the roles of insulin/IGF-I in the development and maintenance of beta-cell mass. Rev Endocr Metab Disord 2005;6:199–210.

    Article  PubMed  CAS  Google Scholar 

  72. Garofalo RS, Orena SJ, Rafidi K, Torchia AJ, Stock JL, Hildebrandt AL, et al. Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKB beta. J Clin Invest 2003;112:197–208.

    PubMed  CAS  Google Scholar 

  73. Nakae J, Biggs WH 3rd, Kitamura T, Cavenee WK, Wright CV, Arden KC, et al. Regulation of insulin action and pancreatic beta-cell function by mutated alleles of the gene encoding forkhead transcription factor Foxo1. Nat Genet 2002;32:245–53.

    Article  PubMed  CAS  Google Scholar 

  74. Tanabe K, Liu Z, Patel S, Doble BW, Li L, Cras-Meneur C, et al. Genetic deficiency of glycogen synthase kinase-3beta corrects diabetes in mouse models of insulin resistance. PLoS Biol 2008;6:e37.

    Article  PubMed  CAS  Google Scholar 

  75. Pende M, Kozma SC, Jaquet M, Oorschot V, Burcelin R, Le Marchand-Brustel Y, et al. Hypoinsulinaemia, glucose intolerance and diminished beta-cell size in S6K1-deficient mice. Nature 2000;408:994–7.

    Article  PubMed  CAS  Google Scholar 

  76. Ruvinsky I, Sharon N, Lerer T, Cohen H, Stolovich-Rain M, Nir T, et al. Ribosomal protein S6 phosphorylation is a determinant of cell size and glucose homeostasis. Genes Dev 2005;19:2199–211.

    Article  PubMed  CAS  Google Scholar 

  77. Rachdi L. BN, Osorio F., Elghazi L., Weiss A., Gould A., Chang-Chen K., et la. Disruption of Tsc2 in pancreatic β-cells induces β-cell mass expansion and improved glucose tolerance in a TORC1-dependent manner. Proc Natl Acad Sci U S A 2008;In Press

  78. Drucker DJ. The biology of incretin hormones. Cell Metab 2006;3:153–65.

    Article  PubMed  CAS  Google Scholar 

  79. Perfetti R, Zhou J, Doyle ME, Egan JM. Glucagon-like peptide-1 induces cell proliferation and pancreatic-duodenum homeobox-1 expression and increases endocrine cell mass in the pancreas of old, glucose-intolerant rats. Endocrinology 2000;141:4600–5.

    Article  PubMed  CAS  Google Scholar 

  80. Stoffers DA, Kieffer TJ, Hussain MA, Drucker DJ, Bonner-Weir S, Habener JF, et al. Insulinotropic glucagon-like peptide 1 agonists stimulate expression of homeodomain protein IDX-1 and increase islet size in mouse pancreas. Diabetes 2000;49:741–8.

    Article  PubMed  CAS  Google Scholar 

  81. De Leon DD, Deng S, Madani R, Ahima RS, Drucker DJ, Stoffers DA. Role of endogenous glucagon-like peptide-1 in islet regeneration after partial pancreatectomy. Diabetes 2003;52:365–71.

    Article  PubMed  Google Scholar 

  82. Tourrel C, Bailbe D, Meile MJ, Kergoat M, Portha B. Glucagon-like peptide-1 and exendin-4 stimulate beta-cell neogenesis in streptozotocin-treated newborn rats resulting in persistently improved glucose homeostasis at adult age. Diabetes 2001;50:1562–70.

    Article  PubMed  CAS  Google Scholar 

  83. Farilla L, Bulotta A, Hirshberg B, Li Calzi S, Khoury N, Noushmehr H, et al. Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology 2003;144:5149–58.

    Article  PubMed  CAS  Google Scholar 

  84. Trumper A, Trumper K, Trusheim H, Arnold R, Goke B, Horsch D. Glucose-dependent insulinotropic polypeptide is a growth factor for beta (INS-1) cells by pleiotropic signaling. Mol Endocrinol 2001;15:1559–70.

    Article  PubMed  CAS  Google Scholar 

  85. Holz GG, Leech CA, Heller RS, Castonguay M, Habener JF. cAMP-dependent mobilization of intracellular Ca2+ stores by activation of ryanodine receptors in pancreatic beta-cells. A Ca2+ signaling system stimulated by the insulinotropic hormone glucagon-like peptide-1-(7–37). J Biol Chem 1999;274:14147–56.

    Article  PubMed  CAS  Google Scholar 

  86. Gromada J, Dissing S, Bokvist K, Renstrom E, Frokjaer-Jensen J, Wulff BS, et al. Glucagon-like peptide I increases cytoplasmic calcium in insulin-secreting beta TC3-cells by enhancement of intracellular calcium mobilization. Diabetes 1995;44:767–74.

    Article  PubMed  CAS  Google Scholar 

  87. Tsuboi T, da Silva Xavier G, Holz GG, Jouaville LS, Thomas AP, Rutter GA. Glucagon-like peptide-1 mobilizes intracellular Ca2+ and stimulates mitochondrial ATP synthesis in pancreatic MIN6 beta-cells. Biochem J 2003;369:287–99.

    Article  PubMed  CAS  Google Scholar 

  88. Wang X, Cahill CM, Pineyro MA, Zhou J, Doyle ME, Egan JM. Glucagon-like peptide-1 regulates the beta cell transcription factor, PDX-1, in insulinoma cells. Endocrinology 1999;140:4904–7.

    Article  PubMed  CAS  Google Scholar 

  89. Li Y, Cao X, Li LX, Brubaker PL, Edlund H, Drucker DJ. beta-Cell Pdx1 expression is essential for the glucoregulatory, proliferative, and cytoprotective actions of glucagon-like peptide-1. Diabetes 2005;54:482–91.

    Article  PubMed  CAS  Google Scholar 

  90. Park S, Dong X, Fisher TL, Dunn SL, Omer AK, Weir G, et al. Exendin-4 promotes IRS2 signaling to mediate pancreatic beta-cell growth and function. J Biol Chem 2005;281:1159–68.

    Article  PubMed  CAS  Google Scholar 

  91. Miettinen PJ, Ustinov J, Ormio P, Gao R, Palgi J, Hakonen E, et al. Downregulation of EGF receptor signaling in pancreatic islets causes diabetes due to impaired postnatal beta-cell growth. Diabetes 2006;55:3299–308.

    Article  PubMed  CAS  Google Scholar 

  92. Buteau J, Foisy S, Joly E, Prentki M. Glucagon-like peptide 1 induces pancreatic beta-cell proliferation via transactivation of the epidermal growth factor receptor. Diabetes 2003;52:124–32.

    Article  PubMed  CAS  Google Scholar 

  93. Poitout V, Robertson RP. Glucolipotoxicity: fuel excess and beta-cell dysfunction. Endocr Rev 2008;29:351–66.

    Article  PubMed  CAS  Google Scholar 

  94. Khaldi MZ, Guiot Y, Gilon P, Henquin JC, Jonas JC. Increased glucose sensitivity of both triggering and amplifying pathways of insulin secretion in rat islets cultured for 1 wk in high glucose. Am J Physiol Endocrinol Metab 2004;287:E207–17.

    Article  PubMed  CAS  Google Scholar 

  95. Finegood DT, McArthur MD, Kojwang D, Thomas MJ, Topp BG, Leonard T, et al. Beta-cell mass dynamics in Zucker diabetic fatty rats. Rosiglitazone prevents the rise in net cell death. Diabetes 2001;50:1021–9.

    Article  PubMed  CAS  Google Scholar 

  96. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 2003;52:102–10.

    Article  PubMed  CAS  Google Scholar 

  97. Olson LK, Redmon JB, Towle HC, Robertson RP. Chronic exposure of HIT cells to high glucose concentrations paradoxically decreases insulin gene transcription and alters binding of insulin gene regulatory protein. J Clin Invest 1993;92:514–9.

    Article  PubMed  CAS  Google Scholar 

  98. Tanaka Y, Gleason CE, Tran PO, Harmon JS, Robertson RP. Prevention of glucose toxicity in HIT-T15 cells and Zucker diabetic fatty rats by antioxidants. Proc Natl Acad Sci U S A 1999;96:10857–62.

    Article  PubMed  CAS  Google Scholar 

  99. Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes. J Clin Invest 2006;116:1802–12.

    Article  PubMed  CAS  Google Scholar 

  100. Tanaka Y, Tran PO, Harmon J, Robertson RP. A role for glutathione peroxidase in protecting pancreatic beta cells against oxidative stress in a model of glucose toxicity. Proc Natl Acad Sci U S A 2002;99:12363–8.

    Article  PubMed  CAS  Google Scholar 

  101. Kaneto H, Kajimoto Y, Miyagawa J, Matsuoka T, Fujitani Y, Umayahara Y, et al. Beneficial effects of antioxidants in diabetes: possible protection of pancreatic beta-cells against glucose toxicity. Diabetes 1999;48:2398–406.

    Article  PubMed  CAS  Google Scholar 

  102. Del Guerra S, Lupi R, Marselli L, Masini M, Bugliani M, Sbrana S, et al. Functional and molecular defects of pancreatic islets in human type 2 diabetes. Diabetes 2005;54:727–35.

    Article  PubMed  CAS  Google Scholar 

  103. Olson LK, Sharma A, Peshavaria M, Wright CV, Towle HC, Rodertson RP, et al. Reduction of insulin gene transcription in HIT-T15 beta cells chronically exposed to a supraphysiologic glucose concentration is associated with loss of STF-1 transcription factor expression. Proc Natl Acad Sci U S A 1995;92:9127–31.

    Article  PubMed  CAS  Google Scholar 

  104. Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-Jemelka HI, Spinas GA, et al. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest 2002;110:851–60.

    PubMed  CAS  Google Scholar 

  105. Kaneto H, Xu G, Fujii N, Kim S, Bonner-Weir S, Weir GC. Involvement of c-Jun N-terminal kinase in oxidative stress-mediated suppression of insulin gene expression. J Biol Chem 2002;277:30010–8.

    Article  PubMed  CAS  Google Scholar 

  106. Aguirre V, Uchida T, Yenush L, Davis R, White MF. The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307). J Biol Chem 2000;275:9047–54.

    Article  PubMed  CAS  Google Scholar 

  107. Aguirre V, Werner ED, Giraud J, Lee YH, Shoelson SE, White MF. Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. J Biol Chem 2002;277:1531–7.

    Article  PubMed  CAS  Google Scholar 

  108. Grill V, Bjorklund A. Overstimulation and beta-cell function. Diabetes 2001;50(Suppl 1):S122–4.

    Article  PubMed  CAS  Google Scholar 

  109. Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, et al. Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature 2004;431:200–5.

    Article  PubMed  CAS  Google Scholar 

  110. Shah OJ, Wang Z, Hunter T. Inappropriate activation of the TSC/Rheb/mTOR/S6K cassette induces IRS1/2 depletion, insulin resistance, and cell survival deficiencies. Curr Biol 2004;14:1650–6.

    Article  PubMed  CAS  Google Scholar 

  111. Harrington LS, Findlay GM, Gray A, Tolkacheva T, Wigfield S, Rebholz H, et al. The TSC1–2 tumor suppressor controls insulin-PI3K signaling via regulation of IRS proteins. J Cell Biol 2004;166:213–23.

    Article  PubMed  CAS  Google Scholar 

  112. Kashyap S, Belfort R, Gastaldelli A, Pratipanawatr T, Berria R, Pratipanawatr W, et al. A sustained increase in plasma free fatty acids impairs insulin secretion in nondiabetic subjects genetically predisposed to develop type 2 diabetes. Diabetes 2003;52:2461–74.

    Article  PubMed  CAS  Google Scholar 

  113. Santomauro AT, Boden G, Silva ME, Rocha DM, Santos RF, Ursich MJ, et al. Overnight lowering of free fatty acids with Acipimox improves insulin resistance and glucose tolerance in obese diabetic and nondiabetic subjects. Diabetes 1999;48:1836–41.

    Article  PubMed  CAS  Google Scholar 

  114. Boden G, Shulman GI. Free fatty acids in obesity and type 2 diabetes: defining their role in the development of insulin resistance and beta-cell dysfunction. Eur J Clin Invest 2002;32(Suppl 3):14–23.

    Article  PubMed  CAS  Google Scholar 

  115. Shimabukuro M, Zhou YT, Levi M, Unger RH. Fatty acid-induced beta cell apoptosis: a link between obesity and diabetes. Proc Natl Acad Sci U S A 1998;95:2498–502.

    Article  PubMed  CAS  Google Scholar 

  116. Prentki M, Joly E, El-Assaad W, Roduit R. Malonyl-CoA signaling, lipid partitioning, and glucolipotoxicity: role in beta-cell adaptation and failure in the etiology of diabetes. Diabetes 2002;51(Suppl 3):S405–13.

    Article  PubMed  CAS  Google Scholar 

  117. El-Assaad W, Buteau J, Peyot ML, Nolan C, Roduit R, Hardy S, et al. Saturated fatty acids synergize with elevated glucose to cause pancreatic beta-cell death. Endocrinology 2003;144:4154–63.

    Article  PubMed  CAS  Google Scholar 

  118. Sako Y, Grill VE. A 48-hour lipid infusion in the rat time-dependently inhibits glucose-induced insulin secretion and B cell oxidation through a process likely coupled to fatty acid oxidation. Endocrinology 1990;127:1580–9.

    Article  PubMed  CAS  Google Scholar 

  119. Elks ML. Chronic perifusion of rat islets with palmitate suppresses glucose-stimulated insulin release. Endocrinology 1993;133:208–14.

    Article  PubMed  CAS  Google Scholar 

  120. Zhou YP, Grill V. Long term exposure to fatty acids and ketones inhibits B-cell functions in human pancreatic islets of Langerhans. J Clin Endocrinol Metab 1995;80:1584–90.

    Article  PubMed  CAS  Google Scholar 

  121. Zhou YP, Grill VE. Long-term exposure of rat pancreatic islets to fatty acids inhibits glucose-induced insulin secretion and biosynthesis through a glucose fatty acid cycle. J Clin Invest 1994;93:870–6.

    Article  PubMed  CAS  Google Scholar 

  122. Gremlich S, Bonny C, Waeber G, Thorens B. Fatty acids decrease IDX-1 expression in rat pancreatic islets and reduce GLUT2, glucokinase, insulin, and somatostatin levels. J Biol Chem 1997;272:30261–9.

    Article  PubMed  CAS  Google Scholar 

  123. Ritz-Laser B, Meda P, Constant I, Klages N, Charollais A, Morales A, et al. Glucose-induced preproinsulin gene expression is inhibited by the free fatty acid palmitate. Endocrinology 1999;140:4005–14.

    Article  PubMed  CAS  Google Scholar 

  124. Jacqueminet S, Briaud I, Rouault C, Reach G, Poitout V. Inhibition of insulin gene expression by long-term exposure of pancreatic beta cells to palmitate is dependent on the presence of a stimulatory glucose concentration. Metabolism 2000;49:532–6.

    Article  PubMed  CAS  Google Scholar 

  125. Briaud I, Harmon JS, Kelpe CL, Segu VB, Poitout V. Lipotoxicity of the pancreatic beta-cell is associated with glucose-dependent esterification of fatty acids into neutral lipids. Diabetes 2001;50:315–21.

    Article  PubMed  CAS  Google Scholar 

  126. Kelpe CL, Moore PC, Parazzoli SD, Wicksteed B, Rhodes CJ, Poitout V. Palmitate inhibition of insulin gene expression is mediated at the transcriptional level via ceramide synthesis. J Biol Chem 2003;278:30015–21.

    Article  PubMed  CAS  Google Scholar 

  127. Hagman DK, Hays LB, Parazzoli SD, Poitout V. Palmitate inhibits insulin gene expression by altering PDX-1 nuclear localization and reducing MafA expression in isolated rat islets of Langerhans. J Biol Chem 2005;280:32413–8.

    Article  PubMed  CAS  Google Scholar 

  128. Pick A, Clark J, Kubstrup C, Levisetti M, Pugh W, Bonner-Weir S, et al. Role of apoptosis in failure of beta-cell mass compensation for insulin resistance and beta-cell defects in the male Zucker diabetic fatty rat. Diabetes 1998;47:358–64.

    Article  PubMed  CAS  Google Scholar 

  129. Cnop M, Hannaert JC, Hoorens A, Eizirik DL, Pipeleers DG. Inverse relationship between cytotoxicity of free fatty acids in pancreatic islet cells and cellular triglyceride accumulation. Diabetes 2001;50:1771–7.

    Article  PubMed  CAS  Google Scholar 

  130. Maedler K, Spinas GA, Dyntar D, Moritz W, Kaiser N, Donath MY. Distinct effects of saturated and monounsaturated fatty acids on beta-cell turnover and function. Diabetes 2001;50:69–76.

    Article  PubMed  CAS  Google Scholar 

  131. Lupi R, Dotta F, Marselli L, Del Guerra S, Masini M, Santangelo C, et al. Prolonged exposure to free fatty acids has cytostatic and pro-apoptotic effects on human pancreatic islets: evidence that beta-cell death is caspase mediated, partially dependent on ceramide pathway, and Bcl-2 regulated. Diabetes 2002;51:1437–42.

    Article  PubMed  CAS  Google Scholar 

  132. Wrede CE, Dickson LM, Lingohr MK, Briaud I, Rhodes CJ. Protein kinase B/Akt prevents fatty acid-induced apoptosis in pancreatic beta-cells (INS-1). J Biol Chem 2002;277:49676–84.

    Article  PubMed  CAS  Google Scholar 

  133. Piro S, Anello M, Di Pietro C, Lizzio MN, Patane G, Rabuazzo AM, et al. Chronic exposure to free fatty acids or high glucose induces apoptosis in rat pancreatic islets: possible role of oxidative stress. Metabolism 2002;51:1340–7.

    Article  PubMed  CAS  Google Scholar 

  134. Maedler K, Oberholzer J, Bucher P, Spinas GA, Donath MY. Monounsaturated fatty acids prevent the deleterious effects of palmitate and high glucose on human pancreatic beta-cell turnover and function. Diabetes 2003;52:726–33.

    Article  PubMed  CAS  Google Scholar 

  135. Maestre I, Jordan J, Calvo S, Reig JA, Cena V, Soria B, et al. Mitochondrial dysfunction is involved in apoptosis induced by serum withdrawal and fatty acids in the beta-cell line INS-1. Endocrinology 2003;144:335–45.

    Article  PubMed  CAS  Google Scholar 

  136. Poitout V, Robertson RP. Minireview: Secondary beta-cell failure in type 2 diabetes–a convergence of glucotoxicity and lipotoxicity. Endocrinology 2002;143:339–42.

    Article  PubMed  CAS  Google Scholar 

  137. Foufelle F, Ferre P. New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c. Biochem J 2002;366:377–91.

    Article  PubMed  CAS  Google Scholar 

  138. Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A. Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic beta-cell apoptosis. Endocrinology 2006;147:3398–407.

    Article  PubMed  CAS  Google Scholar 

  139. Boucher A, Lu D, Burgess SC, Telemaque-Potts S, Jensen MV, Mulder H, et al. Biochemical mechanism of lipid-induced impairment of glucose-stimulated insulin secretion and reversal with a malate analogue. J Biol Chem 2004;279:27263–71.

    Article  PubMed  CAS  Google Scholar 

  140. Joseph JW, Koshkin V, Saleh MC, Sivitz WI, Zhang CY, et al. Free fatty acid-induced beta-cell defects are dependent on uncoupling protein 2 expression. J Biol Chem 2004;279:51049–56.

    Article  PubMed  CAS  Google Scholar 

  141. Moore PC, Ugas MA, Hagman DK, Parazzoli SD, Poitout V. Evidence against the involvement of oxidative stress in fatty acid inhibition of insulin secretion. Diabetes 2004;53:2610–6.

    Article  PubMed  CAS  Google Scholar 

  142. Brunham LR, Kruit JK, Pape TD, Timmins JM, Reuwer AQ, Vasanji Z, et al. Beta-cell ABCA1 influences insulin secretion, glucose homeostasis and response to thiazolidinedione treatment. Nat Med 2007;13:340–7.

    Article  PubMed  CAS  Google Scholar 

  143. Martinez SC, Tanabe K, Cras-Meneur C, Abumrad NA, Bernal-Mizrachi E, Permutt MA. Inhibition of Foxo1 protects pancreatic islet beta-cells against fatty acid and endoplasmic reticulum stress-induced apoptosis. Diabetes 2008;57:846–59.

    Article  PubMed  CAS  Google Scholar 

  144. Robinson KA, Buse MG. Mechanisms of high-glucose/insulin-mediated desensitization of acute insulin-stimulated glucose transport and Akt activation. Am J Physiol Endocrinol Metab 2008;294:E870–81.

    Article  PubMed  CAS  Google Scholar 

  145. Kharroubi I, Ladriere L, Cardozo AK, Dogusan Z, Cnop M, Eizirik DL. Free fatty acids and cytokines induce pancreatic beta-cell apoptosis by different mechanisms: role of nuclear factor-kappaB and endoplasmic reticulum stress. Endocrinology 2004;145:5087–96.

    Article  PubMed  CAS  Google Scholar 

  146. Laybutt DR, Preston AM, Akerfeldt MC, Kench JG, Busch AK, Biankin AV, et al. Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 2007;50:752–63.

    Article  PubMed  CAS  Google Scholar 

  147. Hotamisligil GS. Role of endoplasmic reticulum stress and c-Jun NH2-terminal kinase pathways in inflammation and origin of obesity and diabetes. Diabetes 2005;54(Suppl 2):S73–8.

    Article  PubMed  CAS  Google Scholar 

  148. Nakatani Y, Kaneto H, Kawamori D, Yoshiuchi K, Hatazaki M, Matsuoka TA, et al. Involvement of endoplasmic reticulum stress in insulin resistance and diabetes. J Biol Chem 2005;280:847–51.

    PubMed  CAS  Google Scholar 

  149. Huang CJ, Lin CY, Haataja L, Gurlo T, Butler AE, Rizza RA, et al. High expression rates of human islet amyloid polypeptide induce endoplasmic reticulum stress mediated beta-cell apoptosis, a characteristic of humans with type 2 but not type 1 diabetes. Diabetes 2007;56:2016–27.

    Article  PubMed  CAS  Google Scholar 

  150. Marchetti P, Bugliani M, Lupi R, Marselli L, Masini M, Boggi U, et al. The endoplasmic reticulum in pancreatic beta cells of type 2 diabetes patients. Diabetologia 2007;50:2486–94.

    Article  PubMed  CAS  Google Scholar 

  151. Wang J, Takeuchi T, Tanaka S, Kubo SK, Kayo T, Lu D, et al. A mutation in the insulin 2 gene induces diabetes with severe pancreatic beta-cell dysfunction in the Mody mouse. J Clin Invest 1999;103:27–37.

    Article  PubMed  CAS  Google Scholar 

  152. Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum stress in diabetes mellitus. Endocr Rev 2008;29:42–61.

    Article  PubMed  CAS  Google Scholar 

  153. Kaufman RJ, Scheuner D, Schroder M, Shen X, Lee K, Liu CY, et al. The unfolded protein response in nutrient sensing and differentiation. Nat Rev Mol Cell Biol 2002;3:411–21.

    Article  PubMed  CAS  Google Scholar 

  154. Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol 2000;2:326–32.

    Article  PubMed  CAS  Google Scholar 

  155. Lipson K, Fonseca S, Ishigaki S, Nguyen L, Foss E, Bortell R, et al. Regulation of insulin biosynthesis in pancreatic beta cells by anendoplasmic reticulum-resident protein kinase IRE1. Cell Metabolism 2006;4:245–54.

    Article  PubMed  CAS  Google Scholar 

  156. Lipson KL, Ghosh R, Urano F. The role of IRE1alpha in the degradation of insulin mRNA in pancreatic beta-cells. PLoS ONE 2008;3:e1648.

    Article  PubMed  CAS  Google Scholar 

  157. Pirot P, Naamane N, Libert F, Magnusson NE, Orntoft TF, Cardozo AK, et al. Global profiling of genes modified by endoplasmic reticulum stress in pancreatic beta cells reveals the early degradation of insulin mRNAs. Diabetologia 2007;50:1006–14.

    Article  PubMed  CAS  Google Scholar 

  158. Yoneda T, Imaizumi K, Oono K, Yui D, Gomi F, Katayama T, et al. Activation of caspase-12, an endoplastic reticulum (ER) resident caspase, through tumor necrosis factor receptor-associated factor 2-dependent mechanism in response to the ER stress. J Biol Chem 2001;276:13935–40.

    PubMed  CAS  Google Scholar 

  159. Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000;287:664–6.

    Article  PubMed  CAS  Google Scholar 

  160. Cunha DA, Hekerman P, Ladriere L, Bazarra-Castro A, Ortis F, Wakeham MC, et al. Initiation and execution of lipotoxic ER stress in pancreatic {beta}-cells. J Cell Sci 2008;121:2308–18.

    Article  PubMed  CAS  Google Scholar 

  161. Yoshida H, Haze K, Yanagi H, Yura T, Mori K. Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. J Biol Chem 1998;273:33741–9.

    Article  PubMed  CAS  Google Scholar 

  162. Yamamoto K, Yoshida H, Kokame K, Kaufman RJ, Mori K. Differential contributions of ATF6 and XBP1 to the activation of endoplasmic reticulum stress-responsive cis-acting elements ERSE, UPRE and ERSE-II. J Biochem 2004;136:343–50.

    Article  PubMed  CAS  Google Scholar 

  163. Okada T, Yoshida H, Akazawa R, Negishi M, Mori K. Distinct roles of activating transcription factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase (PERK) in transcription during the mammalian unfolded protein response. Biochem J 2002;366:585–94.

    Article  PubMed  CAS  Google Scholar 

  164. Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, et al. IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response. Genes Dev 2002;16:452–66.

    Article  PubMed  CAS  Google Scholar 

  165. Wu J, Rutkowski DT, Dubois M, Swathirajan J, Saunders T, Wang J, et al. ATF6alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress. Dev Cell 2007;13:351–64.

    Article  PubMed  CAS  Google Scholar 

  166. Thameem F, Farook VS, Bogardus C, Prochazka M. Association of amino acid variants in the activating transcription factor 6 gene (ATF6) on 1q21-q23 with type 2 diabetes in Pima Indians. Diabetes 2006;55:839–42.

    Article  PubMed  CAS  Google Scholar 

  167. Meex SJ, van Greevenbroek MM, Ayoubi TA, Vlietinck R, van Vliet-Ostaptchouk JV, Hofker MH, et al. Activating transcription factor 6 polymorphisms and haplotypes are associated with impaired glucose homeostasis and type 2 diabetes in Dutch Caucasians. J Clin Endocrinol Metab 2007;92:2720–5.

    Article  PubMed  CAS  Google Scholar 

  168. Seo HY, Kim YD, Lee KM, Min AK, Kim MK, Kim HS, et al. ER stress-induced Activation of ATF6 Decreases Insulin Gene Expression via Upregulation of Orphan Nuclear Receptor SHP. Endocrinology. 2008;149–3832–41.

  169. Hinnebusch AG. Gene-specific translational control of the yeast GCN4 gene by phosphorylation of eukaryotic initiation factor 2. Mol Microbiol 1993;10:215–23.

    Article  PubMed  CAS  Google Scholar 

  170. Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M, et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell 2000;6:1099–108.

    Article  PubMed  CAS  Google Scholar 

  171. Vattem KM, Wek RC. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc Natl Acad Sci U S A 2004;101:11269–74.

    Article  PubMed  CAS  Google Scholar 

  172. Wek RC, Cavener DR. Translational control and the unfolded protein response. Antioxid Redox Signal 2007;9:2357–71.

    Article  PubMed  CAS  Google Scholar 

  173. Tajiri S, Oyadomari S, Yano S, Morioka M, Gotoh T, Hamada JI, et al. Ischemia-induced neuronal cell death is mediated by the endoplasmic reticulum stress pathway involving CHOP. Cell Death Differ 2004;11:403–15.

    Article  PubMed  CAS  Google Scholar 

  174. Ma Y, Brewer JW, Diehl JA, Hendershot LM. Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J Mol Biol 2002;318:1351–65.

    Article  PubMed  CAS  Google Scholar 

  175. Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, Sadri N, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 2003;11:619–33.

    Article  PubMed  CAS  Google Scholar 

  176. Yamaguchi S, Ishihara H, Yamada T, Tamura A, Usui M, Tominaga R, et al. ATF4-mediated induction of 4E-BP1 contributes to pancreatic beta cell survival under endoplasmic reticulum stress. Cell Metab 2008;7:269–76.

    Article  PubMed  CAS  Google Scholar 

  177. Cnop M, Ladriere L, Hekerman P, Ortis F, Cardozo AK, Dogusan Z, et al. Selective inhibition of eukaryotic translation initiation factor 2 alpha dephosphorylation potentiates fatty acid-induced endoplasmic reticulum stress and causes pancreatic beta-cell dysfunction and apoptosis. J Biol Chem 2007;282:3989–97.

    Article  PubMed  CAS  Google Scholar 

  178. Pirot P, Ortis F, Cnop M, Ma Y, Hendershot LM, Eizirik DL, et al. Transcriptional regulation of the endoplasmic reticulum stress gene chop in pancreatic insulin-producing cells. Diabetes 2007;56:1069–77.

    Article  PubMed  CAS  Google Scholar 

  179. Harding H, Zeng H, Zhang Y, Jungreis R, Chung P, Plesken H, et al. Diabetes Mellitus and Exocrine Pancreatic Dysfunction in Perk/Mice Reveals a Role for Translational Control in Secretory Cell Survival. Mol Cell 2001;7:1153–63.

    Article  PubMed  CAS  Google Scholar 

  180. Oyadomari S, Koizumi A, Takeda K, Gotoh T, Akira S, Araki E, et al. Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes. J Clin Invest 2002;109:525–32.

    PubMed  CAS  Google Scholar 

  181. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 2004;306:457–61.

    Article  PubMed  CAS  Google Scholar 

  182. Li D, Yin X, Zmuda EJ, Wolford CC, Dong X, White MF, et al. The repression of IRS2 gene by ATF3, a stress-inducible gene, contributes to pancreatic beta-cell apoptosis. Diabetes 2008;57:635–44.

    Article  PubMed  CAS  Google Scholar 

  183. Wang H, Kouri G, Wollheim CB. ER stress and SREBP-1 activation are implicated in beta -cell glucolipotoxicity. J Cell Sci 2005;118:3905–15.

    Article  PubMed  CAS  Google Scholar 

  184. Kawamori D, Kajimoto Y, Kaneto H, Umayahara Y, Fujitani Y, Miyatsuka T, et al. Oxidative stress induces nucleo-cytoplasmic translocation of pancreatic transcription factor PDX-1 through activation of c-Jun NH(2)-terminal kinase. Diabetes 2003;52:2896–904.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to E. Bernal-Mizrachi.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chang-Chen, K.J., Mullur, R. & Bernal-Mizrachi, E. β-cell failure as a complication of diabetes. Rev Endocr Metab Disord 9, 329–343 (2008). https://doi.org/10.1007/s11154-008-9101-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11154-008-9101-5

Keywords

Navigation