Skip to main content
Log in

Endoplasmic reticulum stress-mediated apoptosis in pancreatic β-cells

  • Published:
Apoptosis Aims and scope Submit manuscript

Abstract

Apoptotic cell death in pancreatic β-cells is involved in the pathogenesis of diabetes. Signals from death receptors and DNA damage have been widely accepted as being triggers of apoptosis in β-cells. Recent studies indicated that the endoplasmic reticulum (ER) can sense and transduce apoptotic signals. Various genetic and environmental stresses interfere with protein folding in the ER and induce ER stress. In mammals, ER stress transducer proteins IRE1, PERK and ATF6 activate both survival and apoptotic pathways. The former includes transcriptional induction of ER chaperones, translational attenuation, and ER-associated degradation (ERAD) while the latter includes transcriptional induction of CHOP/GADD153, the activation of cJUN NH2-terminal kinase, and the activation of caspase-12. A characteristic feature of β-cells is the highly developed ER apparently due to a heavy engagement in insulin secretion. β-cells are most susceptible to ER stress. The recent studies reviewed in this article revealed that ER stress-mediated apoptosis in β-cells plays an important role in the development of diabetes.

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.

Institutional subscriptions

Similar content being viewed by others

References

  1. Bonner-Weir S Perspective: Postnatal pancreatic β cell growth. Endocrinology 2000; 141: 1926-1929.

    Google Scholar 

  2. Mathis D, Vence L, Benoist C ββ-Cell death during progression to diabetes. Nature 2001; 414: 792-798.

    Google Scholar 

  3. Eizirik DL, Mandrup-Poulsen T A choice of death—the signal—transduction of immune-mediated β-cell apoptosis. Diabetologia 2001; 44: 2115-2133.

    Google Scholar 

  4. Mori K Tripartite management of unfolded proteins in the endoplasmic reticulum. Cell 2000; 101: 451-454.

    Google Scholar 

  5. Kaufman RJ Stress signaling from the lumen of the endoplasmic reticulum: Coordination of gene transcriptional and translational controls. Genes Dev 1999; 13: 1211-1233.

    Google Scholar 

  6. 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: 33741-33749.

    Google Scholar 

  7. Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 2001; 107: 881-891.

    Google Scholar 

  8. Shen X, Ellis RE, Lee K, et al Complementary signaling pathways regulate the unfolded protein response and are required for C. elegans development. Cell 2001; 107: 893-903.

    Google Scholar 

  9. Calfon M, Zeng H, Urano F, et al IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 2002; 415: 92-96.

    Google Scholar 

  10. Yoshida H, Okada T, Haze K, et al ATF6 activated by proteolysis binds in the presence of NF-Y (CBF) directly to the cis-acting element responsible for the mammalian unfolded protein response. Mol Cell Biol 2000; 20: 6755-6767.

    Google Scholar 

  11. Chen X, Shen J, Prywes R The lumenal domain of ATF6 senses ER stress and causes translocation of ATF6 from the ER to the Golgi. J Biol Chem 2002; 30: 30.

    Google Scholar 

  12. Ye J, Rawson RB, Komuro R, et al ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell 2000; 6: 1355-1364.

    Google Scholar 

  13. 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: 326-332.

    Google Scholar 

  14. Shi Y, Vattem KM, Sood R, et al Identification and characterization of pancreatic eukaryotic initiation factor 2 α-subunit kinase, PEK, involved in translational control. Mol Cell Biol 1998; 18: 7499-7509.

    Google Scholar 

  15. Harding HP, Zhang Y, Ron D Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 1999; 397: 271-274.

    Google Scholar 

  16. Harding HP, Zhang Y, Bertolotti A, Zeng H, Ron D Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol Cell 2000; 5: 897-904.

    Google Scholar 

  17. Novoa I, Zeng H, Harding HP, Ron D Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2α. J Cell Biol 2001; 153: 1011-1022.

    Google Scholar 

  18. Iwawaki T, Hosoda A, Okuda T, et al Translational control by the ER transmembrane kinase/ribonuclease IRE1 under ER stress. Nat Cell Biol 2001; 3: 158-164.

    Google Scholar 

  19. Jakob CA, Burda P, Roth J, Aebi M Degradation of misfolded endoplasmic reticulum glycoproteins in Saccharomyces cerevisiae is determined by a specific oligosaccharide structure. J Cell Biol 1998; 142: 1223-1233.

    Google Scholar 

  20. Jakob CA, Bodmer D, Spirig U, et al Htm1p, α mannosidaselike protein, is involved in glycoprotein degradation in yeast. EMBO Rep 2001; 2: 423-430.

    Google Scholar 

  21. Nakatsukasa K, Nishikawa S, Hosokawa N, Nagata K, Endo T Mnl1p, an α-mannosidase-like protein in yeast Saccharomyces cerevisiae, is required for endoplasmic reticulumassociated degradation of glycoproteins. J Biol Chem 2001; 276: 8635-8638.

    Google Scholar 

  22. Hosokawa N, Wada I, Hasegawa K, et al A novel ER α-mannosidase-like protein accelerates ER-associated degradation. EMBO Rep 2001; 2: 415-422.

    Google Scholar 

  23. Ellgaard L, Helenius A ER quality control: Towards an understanding at the molecular level. Curr Opin Cell Biol 2001; 13: 431-437.

    Google Scholar 

  24. Nishikawa SI, Fewell SW, Kato Y, Brodsky JL, Endo T Molecular chaperones in the yeast endoplasmic reticulum maintain the solubility of proteins for retrotranslocation and degradation. J Cell Biol 2001; 153: 1061-1070.

    Google Scholar 

  25. Molinari M, Helenius A Chaperone selection during glycoprotein translocation into the endoplasmic reticulum. Science 2000; 288: 331-333.

    Google Scholar 

  26. Walter J, Urban J, Volkwein C, Sommer T Sec61pindependent degradation of the tail-anchored ER membrane protein Ubc6p. EMBO J 2001; 20: 3124-3131.

    Google Scholar 

  27. Hiller MM, Finger A, Schweiger M, Wolf DH. ER degradation of a misfolded luminal protein by the cytosolic ubiquitinproteasome pathway. Science 1996; 273: 1725-1728.

    Google Scholar 

  28. Biederer T, Volkwein C, Sommer T Degradation of subunits of the Sec61p complex, an integral component of the ER membrane, by the ubiquitin-proteasome pathway. EMBO J 1996; 15: 2069-2076.

    Google Scholar 

  29. Knop M, Finger A, Braun T, Hellmuth K, Wolf DH Der1, a novel protein specifically required for endoplasmic reticulum degradation in yeast. EMBO J 1996; 15: 753-763.

    Google Scholar 

  30. Hampton RY, Gardner RG, Rine J Role of 26S proteasome and HRD genes in the degradation of 3-hydroxy-3-methylglutaryl-CoA reductase, an integral endoplasmic reticulum membrane protein. Mol Biol Cell 1996; 7: 2029-2044.

    Google Scholar 

  31. Bordallo J, Plemper RK, Finger A, Wolf DH Der3p/Hrd1p is required for endoplasmic reticulum-associated degradation of misfolded lumenal and integral membrane proteins. Mol Biol Cell 1998; 9: 209-222.

    Google Scholar 

  32. Casagrande R, Stern P, Diehn M, et al Degradation of proteins from the ER of S. cerevisiae requires an intact unfolded protein response pathway. Mol Cell 2000; 5: 729-735.

    Google Scholar 

  33. Friedlander R, Jarosch E, Urban J, Volkwein C, Sommer T A regulatory link between ER-associated protein degradation and the unfolded-protein response. Nat Cell Biol 2000; 2: 379-384.

    Google Scholar 

  34. Travers KJ, Patil CK, Wodicka L, Lockhart DJ, Weissman JS, Walter P Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ERassociated degradation. Cell 2000; 101: 249-258.

    Google Scholar 

  35. Ron D, Habener JF CHOP, a novel developmentally regulated nuclear protein that dimerizes with transcription factors C/EBP and LAP and functions as a dominant-negative inhibitor of gene transcription. Genes Dev 1992; 6: 439-453.

    Google Scholar 

  36. Wang XZ, Lawson B, Brewer JW, et al Signals from the stressed endoplasmic reticulum induce C/EBP-homologous protein (CHOP/GADD153). Mol Cell Biol 1996; 16: 4273-4280.

    Google Scholar 

  37. Barone MV, Crozat A, Tabaee A, Philipson L, Ron D CHOP (GADD153) and its oncogenic variant, TLS-CHOP, have opposing effects on the induction of G1/S arrest. Genes Dev 1994; 8: 453-464.

    Google Scholar 

  38. Matsumoto M, Minami M, Takeda K, Sakao Y, Akira S Ectopic expression of CHOP (GADD153) induces apoptosis in M1 myeloblastic leukemia cells. FEBS Lett 1996; 395: 143-147.

    Google Scholar 

  39. Gotoh T, Oyadomari S, Mori K, Mori M Nitric oxide-induced apoptosis in RAW 264.7 macrophages is mediated by endoplasmic reticulum stress pathway involvingATF6 andCHOP. J Biol Chem 2002; 22: 22.

    Google Scholar 

  40. Zinszner H, Kuroda M, Wang X, et al CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev 1998; 12: 982-995.

    Google Scholar 

  41. Oyadomari S, Takeda K, Takiguchi M, et al Nitric oxideinduced apoptosis in pancreatic β cells is mediated by the endoplasmic reticulum stress pathway. Proc Natl Acad Sci USA 2001; 98: 10845-10850.

    Google Scholar 

  42. Oyadomari S, Koizumi A, Takeda K, et al Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes. J Clin Invest 2002; 109: 525-532.

    Google Scholar 

  43. Wang XZ, Harding HP, Zhang Y, Jolicoeur EM, Kuroda M, Ron D Cloning of mammalian Ire1 reveals diversity in the ER stress responses. EMBO J 1998; 17: 5708-5717.

    Google Scholar 

  44. Harding HP, Novoa II, Zhang Y, et al Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell 2000; 6: 1099-1108.

    Google Scholar 

  45. Wang XZ, Ron D Stress-induced phosphorylation and activation of the transcription factor CHOP (GADD153) by p38 MAP Kinase. Science 1996; 272: 1347-1349.

    Google Scholar 

  46. Chen BP, Wolfgang CD, Hai T Analysis of ATF3, a transcription factor induced by physiological stresses and modulated by gadd153/Chop10. Mol Cell Biol 1996; 16: 1157-1168.

    Google Scholar 

  47. Ubeda M, Vallejo M, Habener JF CHOP enhancement of gene transcription by interactions with Jun/Fos AP-1 complex proteins. Mol Cell Biol 1999; 19: 7589-7599.

    Google Scholar 

  48. Wang XZ, Kuroda M, Sok J, et al Identification of novel stress-induced genes downstream of chop. EMBO J 1998; 17: 3619-3630.

    Google Scholar 

  49. Sok J, Wang XZ, Batchvarova N, Kuroda M, Harding H, Ron D CHOP-Dependent stress-inducible expression of a novel form of carbonic anhydrase VI. Mol Cell Biol 1999; 19: 495-504.

    Google Scholar 

  50. Antonsson B, Conti F, Ciavatta A, et al Inhibition of Bax channel-forming activity by Bcl-2. Science 1997; 277: 370-372.

    Google Scholar 

  51. Wei MC, Zong WX, Cheng EH, et al Proapoptotic BAX and BAK: A requisite gateway to mitochondrial dysfunction and death. Science 2001; 292: 727-730.

    Google Scholar 

  52. McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol 2001; 21: 1249-1259.

    Google Scholar 

  53. Urano F, Wang X, Bertolotti A, et al Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000; 287: 664-666.

    Google Scholar 

  54. Tobiume K, Matsuzawa A, Takahashi T, et al ASK1 is required for sustained activations of JNK/p38 MAP kinases and apoptosis. EMBO Rep 2001; 2: 222-228.

    Google Scholar 

  55. Nakagawa T, Zhu H, Morishima N, et al Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-β. Nature 2000; 403: 98-103.

    Google Scholar 

  56. Nakagawa T, Yuan J Cross-talk between two cysteine protease families. Activation of caspase-12 by calpain in apoptosis. J Cell Biol 2000; 150: 887-894.

    Google Scholar 

  57. Yoneda T, Imaizumi K, Oono K, 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: 13935-13940.

    Google Scholar 

  58. Rao RV, Hermel E, Castro-Obregon S, et al Coupling endoplasmic reticulum stress to the cell death program. Mechanism of caspase activation. J Biol Chem 2001; 276: 33869-33874.

    Google Scholar 

  59. Chandler JM, Cohen GM, MacFarlane M Different subcellular distribution of caspase-3 and caspase-7 following Fasinduced apoptosis in mouse liver. J Biol Chem 1998; 273: 10815-10818.

    Google Scholar 

  60. Mauricio D, Mandrup-Poulsen T Apoptosis and the pathogenesis of IDDM: A question of life and death. Diabetes 1998; 47: 1537-1543.

    Google Scholar 

  61. Ignarro LJ Introduction and overview. In: Ignarro LJ, ed. Nitric Oxide Biology and Pathobiology. San Diego: Academic Press, 2000: 3-19.

    Google Scholar 

  62. Messmer UK, Ankarcrona M, Nicotera P, Brune B p53 expression in nitric oxide-induced apoptosis. FEBS Lett 1994; 355: 23-26.

    Google Scholar 

  63. Pieper AA, Verma A, Zhang J, Snyder SH Poly (ADP-ribose) polymerase, nitric oxide and cell death. Trends Pharmacol Sci 1999; 20: 171-181.

    Google Scholar 

  64. Enoch T, Norbury C Cellular responses to DNA damage: Cell-cycle checkpoints, apoptosis and the roles of p53 and ATM. Trends Biochem Sci 1995; 20: 426-430.

    Google Scholar 

  65. Messmer UK, Brune B Nitric oxide-induced apoptosis: p53-dependent and p53-independent signalling pathways. Biochem J 1996; 319: 299-305.

    Google Scholar 

  66. Heller B, Wang ZQ, Wagner EF, et al Inactivation of the poly(ADP-ribose) polymerase gene affects oxygen radical and nitric oxide toxicity in islet cells. J Biol Chem 1995; 270: 11176-11180.

    Google Scholar 

  67. Kawahara K, Oyadomari S, Gotoh T, Kohsaka S, Nakayama H, Mori M Induction of CHOP and apoptosis by nitric oxide in p53-deficient microglial cells. FEBS Lett 2001; 506: 135-139.

    Google Scholar 

  68. Doutheil J, Althausen S, Treiman M, Paschen W Effect of nitric oxide on endoplasmic reticulum calcium homeostasis, protein synthesis and energy metabolism. Cell Calcium 2000; 27: 107-115.

    Google Scholar 

  69. Cardozo AK, Kruhoffer M, Leeman R, Orntoft T, Eizirik DL Identification of novel cytokine-induced genes in pancreatic β-cells by high-density oligonucleotide arrays. Diabetes 2001; 50: 909-920.

    Google Scholar 

  70. He H, Lam M, McCormick TS, Distelhorst CW Maintenance of calcium homeostasis in the endoplasmic reticulum by Bcl-2. J Cell Biol 1997; 138: 1219-1228.

    Google Scholar 

  71. Zhou YP, Teng D, Dralyuk F, et al Apoptosis in insulinsecreting cells. Evidence for the role of intracellular Ca2+ stores and arachidonic acid metabolism. J Clin Invest 1998; 101: 1623-1632.

    Google Scholar 

  72. Corbett EF, Michalak M Calcium, a signaling molecule in the endoplasmic reticulum? Trends Biochem Sci 2000; 25: 307-311.

    Google Scholar 

  73. Varadi A, Molnar E, Ostenson CG, Ashcroft SJ Isoforms of endoplasmic reticulum Ca2+-ATPase are differentially expressed in normal and diabetic islets of Langerhans. Biochem J 1996; 319: 521-527.

    Google Scholar 

  74. Viner RI, Ferrington DA, Williams TD, Bigelow DJ, Schoneich C Protein modification during biological aging: Selective tyrosine nitration of the SERCA2a isoform of the sarcoplasmic reticulum Ca2+-ATPase in skeletal muscle. Biochem J 1999; 340: 657-669.

    Google Scholar 

  75. Xu KY, Huso DL, Dawson TM, Bredt DS, Becker LC Nitric oxide synthase in cardiac sarcoplasmic reticulum. Proc Natl Acad Sci USA 1999; 96: 657-662.

    Google Scholar 

  76. Caspersen C, Pedersen PS, Treiman M The sarco/endoplasmic reticulum calcium-ATPase 2b is an endoplasmic reticulum stress-inducible protein. J Biol Chem 2000; 275: 22363-22372.

    Google Scholar 

  77. Blondel O, Takeda J, Janssen H, Seino S, Bell GI Sequence and functional characterization of a third inositol trisphosphate receptor subtype, IP3R-3, expressed in pancreatic islets, kidney, gastrointestinal tract, and other tissues. J Biol Chem 1993; 268: 11356-11363.

    Google Scholar 

  78. Schlossmann J, Ammendola A, Ashman K, et al Regulation of intracellular calcium by a signalling complex of IRAG, IP3 receptor and cGMP kinase Iβ. Nature 2000; 404: 197-201.

    Google Scholar 

  79. Xu L, Eu JP, Meissner G, Stamler JS Activation of the cardiac calcium release channel (ryanodine receptor) by poly-Snitrosylation. Science 1998; 279: 234-237.

    Google Scholar 

  80. Islam MS, Leibiger I, Leibiger B, et al In situ activation of the type 2 ryanodine receptor in pancreatic β cells requires cAMP-dependent phosphorylation. Proc Natl Acad Sci USA 1998; 95: 6145-6150.

    Google Scholar 

  81. DeFronzo RA, Ferrannini E Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care 1991; 14: 173-194.

    Google Scholar 

  82. Stefan Y, Orci L, Malaisse-Lagae F, Perrelet A, Patel Y, Unger RH Quantitation of endocrine cell content in the pancreas of nondiabetic and diabetic humans. Diabetes 1982; 31: 694-700.

    Google Scholar 

  83. Rahier J, Goebbels RM, Henquin JC Cellular composition of the human diabetic pancreas. Diabetologia 1983; 24: 366-371.

    Google Scholar 

  84. Yoshioka M, Kayo T, Ikeda T, Koizumi A A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes 1997; 46: 887-894.

    Google Scholar 

  85. Kayo T, Koizumi A Mapping of murine diabetogenic gene mody on chromosome 7 at D7Mit258 and its involvement in pancreatic islet and β cell development during the perinatal period. J Clin Invest 1998; 101: 2112-2118.

    Google Scholar 

  86. Wang J, Takeuchi T, Tanaka S, et al A mutation in the insulin 2 gene induces diabetes with severe pancreatic β-cell dysfunction in the Mody mouse. J Clin Invest 1999; 103: 27-37.

    Google Scholar 

  87. Leroux L, Desbois P, Lamotte L, et al Compensatory responses in mice carrying a null mutation for Ins1 or Ins2. Diabetes 2001; 50 Suppl 1: S150-153.

    Google Scholar 

  88. Harding HP, Zeng H, Zhang Y, 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: 1153-1163.

    Google Scholar 

  89. Scheuner D, Song B, McEwen E, et al Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 2001; 7: 1165-1176.

    Google Scholar 

  90. Delepine M, Nicolino M, Barrett T, Golamaully M, Lathrop GM, Julier C EIF2AK3, encoding translation initiation factor 2-α kinase 3, is mutated in patients withWolcott-Rallison syndrome. Nat Genet 2000; 25: 406-409.

    Google Scholar 

  91. Thornton CM, Carson DJ, Stewart FJ Autopsy findings in the Wolcott-Rallison syndrome. Pediatr Pathol Lab Med 1997; 17: 487-496.

    Google Scholar 

  92. Kaneto H, Kajimoto Y, Miyagawa J, et al Beneficial effects of antioxidants in diabetes: Possible protection of pancreatic β-cells against glucose toxicity. Diabetes 1999; 48: 2398-2406.

    Google Scholar 

  93. Saini KS, Thompson C, Winterford CM, Walker NI, Cameron DP Streptozotocin at low doses induces apoptosis and at high doses causes necrosis in a murine pancreatic β cell line, INS-1. Biochem Mol Biol Int 1996; 39: 1229-1236.

    Google Scholar 

  94. Delaney CA, Dunger A, Di Matteo M, Cunningham JM, Green MH, Green IC Comparison of inhibition of glucosestimulated insulin secretion in rat islets of Langerhans by streptozotocin and methyl and ethyl nitrosoureas and methanesulphonates. Lack of correlation with nitric oxidereleasing or O6-alkylating ability. Biochem Pharmacol 1995; 50: 2015-2020.

    Google Scholar 

  95. Unger RH Lipotoxic diseases. Annu Rev Med 2002; 53: 319-336.

    Google Scholar 

  96. Withers DJ, Burks DJ, Towery HH, Altamuro SL, Flint CL, White MF Irs-2 coordinates Igf-1 receptor-mediated β-cell development and peripheral insulin signalling. Nat Genet 1999; 23: 32-40.

    Google Scholar 

  97. Withers DJ, Gutierrez JS, Towery H, et al Disruption of IRS-2 causes type 2 diabetes in mice. Nature 1998; 391: 900-904.

    Google Scholar 

  98. Miettinen PJ, Huotari M, Koivisto T, et al Impaired migration and delayed differentiation of pancreatic islet cells in mice lacking EGF-receptors. Development 2000; 127: 2617-2627.

    Google Scholar 

  99. Hart AW, Baeza N, Apelqvist A, Edlund H Attenuation of FGF signalling in mouse β-cells leads to diabetes. Nature 2000; 408: 864-868.

    Google Scholar 

  100. Liu K, Paterson AJ, Chin E, Kudlow JE Glucose stimulates protein modification by O-linked GlcNAc in pancreatic β cells: Linkage of O-linked GlcNAc to β cell death. Proc Natl Acad Sci USA 2000; 97: 2820-2825.

    Google Scholar 

  101. Biarnes M, Montolio M, Nacher V, Raurell M, Soler J, Montanya E β-cell death and mass in syngeneically transplanted islets exposed to short-and long-term hyperglycemia. Diabetes 2002; 51: 66-72.

    Google Scholar 

  102. Zhang S, Liu J, Saafi EL, Cooper GJ Induction of apoptosis by human amylin in RINm5F islet β-cells is associated with enhanced expression of p53 and p21WAF1/CIP1. FEBS Lett 1999; 455: 315-320.

    Google Scholar 

  103. Rane SG, Dubus P, Mettus RV, 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.

    Google Scholar 

  104. Naya FJ, Huang HP, Qiu Y, et al Diabetes, defective pancreatic morphogenesis, and abnormal enteroendocrine differentiation in BETA2/neuroD-deficient mice. Genes Dev 1997; 11: 2323-2334.

    Google Scholar 

  105. Wobser H, Dussmann H, Kogel D, et al Dominant-negative suppression of HNF-1α results in mitochondrial dysfunction, INS-1 cell apoptosis, and increased sensitivity to ceramide-, but not to high glucose-induced cell death. J Biol Chem 2002; 277: 6413-6421.

    Google Scholar 

  106. Silva JP, Kohler M, Graff C, et al Impaired insulin secretion and β-cell loss in tissue-specific knockout mice with mitochondrial diabetes. Nat Genet 2000; 26: 336-340.

    Google Scholar 

  107. Nishitoh H, Matsuzawa A, Tobiume K, et al ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev in press.

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Oyadomari, S., Araki, E. & Mori, M. Endoplasmic reticulum stress-mediated apoptosis in pancreatic β-cells. Apoptosis 7, 335–345 (2002). https://doi.org/10.1023/A:1016175429877

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1016175429877

Navigation