Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-7drxs Total loading time: 0 Render date: 2024-07-17T14:39:56.677Z Has data issue: false hasContentIssue false

19 - Contribution of Apoptosis to Physiologic Remodeling of the Endocrine Pancreas and Pathophysiology of Diabetes

from Part II - Cell Death in Tissues and Organs

Published online by Cambridge University Press:  07 September 2011

Douglas R. Green
Affiliation:
St. Jude Children's Research Hospital, Memphis, Tennessee
Nika N. Danial
Affiliation:
Dana-Farber Cancer Institute
John C. Reed
Affiliation:
Sanford-Burnham Medical Research Institute, La Jolla, California
Get access

Summary

Introduction

Homeostatic control of blood glucose levels is critically dependent on the balance of glucagon and insulin, two counteracting pancreatic hormones secreted by endocrine cells within the islets of Langerhans – alpha and beta cells, respectively. Elegant biochemical studies combined with metabolic flux analysis uncovered the unique ability of beta cells to sense blood glucose fluctuations and to fine tune insulin secretion accordingly. A high-capacity glucose transport system, a low-Km glucose phosphorylating activity catalyzed by glucokinase (GK), and the ability to channel the majority of glycolytically derived pyruvate to the mitochondrial tricarboxylic acid (TCA) cycle constitute essential metabolic design features that endow beta cells with a specialized secretory function. The increase in intracellular adenosine triphosphate (ATP)/adenosine diphosphate (ADP) ratio on mitochondrial metabolism of nutrients is among the metabolic coupling factors connecting fuel oxidation to insulin secretion. A rise in ATP/ADP ratio in turn leads to closure of ATP-sensitive K (KATP) channels at the plasma membrane, followed by membrane depolarization and influx of Ca+ necessary for release of insulin granules. The two aspects of beta cell biology that contribute significantly to euglycemia are glucose dose responsiveness of insulin secretion and the remarkable plasticity of beta cell mass to meet insulin demand under physiologic and pathophysiologic nutrient stress. Beta cell mass is the net outcome of neogenesis (formation of new beta cells from non–beta cell precursors), replication of preexisting beta cells, beta cell size, and apoptosis. The integration of beta cell function and mass is further ensured through nutrient sensing pathways that concomitantly signal insulin secretion and modulate beta cell replication and survival. This chapter highlights the apoptotic mechanisms operative in beta cells that influence the dynamic control of beta cell mass during the developmental remodeling of the endocrine pancreas and in the pathogenesis of diabetes.

Type
Chapter
Information
Apoptosis
Physiology and Pathology
, pp. 201 - 220
Publisher: Cambridge University Press
Print publication year: 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Matschinsky, F.M.Lecture, Banting 1995. A lesson in metabolic regulation inspired by the glucokinase glucose sensor paradigm. Diabetes 45, 223–41 (1996).
Wiederkehr, A. & Wollheim, C.B. Minireview: implication of mitochondria in insulin secretion and action. Endocrinology 147, 2643–9 (2006).
Rorsman, P. The pancreatic beta-cell as a fuel sensor: an electrophysiologist's viewpoint. Diabetologia 40, 487–95 (1997).
Bonner-Weir, S. Life and death of the pancreatic beta cells. Trends Endocrinol Metab 11, 375–8 (2000).
Bouwens, L. & Rooman, I. Regulation of pancreatic beta-cell mass. Physiol Rev 85, 1255–70 (2005).
Bonner-Weir, S. Perspective: Postnatal pancreatic beta cell growth. Endocrinology 141, 1926–9 (2000).
Brubaker, P.L. & Drucker, D.J. Minireview: Glucagon-like peptides regulate cell proliferation and apoptosis in the pancreas, gut, and central nervous system. Endocrinology 145, 2653–9 (2004).
de Koning, E.J., Bonner-Weir, S. & Rabelink, T.J. Preservation of beta-cell function by targeting beta-cell mass. Trends Pharmacol Sci 29, 218–27 (2008).
McEvoy, R.C. & Madson, K.L. Pancreatic insulin-, glucagon-, and somatostatin-positive islet cell populations during the perinatal development of the rat. I. Morphometric quantitation. Biol Neonate 38, 248–54 (1980).
Stefan, Y., Grasso, S., Perrelet, A. & Orci, L. A quantitative immunofluorescent study of the endocrine cell populations in the developing human pancreas. Diabetes 32, 293–301 (1983).
Kassem, S.A., Ariel, I., Thornton, P.S., Scheimberg, I. & Glaser, B. Beta-cell proliferation and apoptosis in the developing normal human pancreas and in hyperinsulinism of infancy. Diabetes 49, 1325–33 (2000).
Scaglia, L., Cahill, C.J., Finegood, D.T. & Bonner-Weir, S. Apoptosis participates in the remodeling of the endocrine pancreas in the neonatal rat. Endocrinology 138, 1736–41 (1997).
Freinkel, N.. Differential effects of age versus glycemic stimulation on the maturation of insulin stimulus-secretion coupling during culture of fetal rat islets. Diabetes 33, 1028–38 (1984).
Pipeleers, D., Kiekens, R., Ling, Z., Wilikens, A. & Schuit, F. Physiologic relevance of heterogeneity in the pancreatic beta-cell population. Diabetologia 37 Suppl 2, S57–64 (1994).
Wikstrom, J.D.. Beta-cell mitochondria exhibit membrane potential heterogeneity that can be altered by stimulatory or toxic fuel levels. Diabetes 56, 2569–78 (2007).
Dunne, M.J., Cosgrove, K.E., Shepherd, R.M., Aynsley-Green, A. & Lindley, K.J. Hyperinsulinism in infancy: from basic science to clinical disease. Physiol Rev 84, 239–75 (2004).
Altieri, D.C. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer 8, 61–70 (2008).
Bouwens, L. & De Blay, E. Islet morphogenesis and stem cell markers in rat pancreas. J Histochem Cytochem 44, 947–51 (1996).
Liggins, C., Orlicky, D.J., Bloomquist, L.A. & Gianani, R. Developmentally regulated expression of Survivin in human pancreatic islets. Pediatr Dev Pathol 6, 392–7 (2003).
Datta, S.R.. 14–3-3 proteins and survival kinases cooperate to inactivate BAD by BH3 domain phosphorylation. Mol Cell 6, 41–51. (2000).
Hettiarachchi, K.D., Zimmet, P.Z., Danial, N.N. & Myers, M.A. Transplacental exposure to the vacuolar-ATPase inhibitor bafilomycin disrupts survival signaling in beta cells and delays neonatal remodeling of the endocrine pancreas. Exp Toxicol Pathol (2008).
Zha, J., Harada, H., Yang, E., Jockel, J. & Korsmeyer, S.J. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14–3-3 not BCL-X(L). Cell 87, 619–28. (1996).
Harada, H., Andersen, J.S., Mann, M., Terada, N. & Korsmeyer, S.J. p70S6 kinase signals cell survival as well as growth, inactivating the pro-apoptotic molecule BAD. Proc Natl Acad Sci U S A 98, 9666–70. (2001).
Petrik, J., Arany, E., McDonald, T.J. & Hill, D.J. Apoptosis in the pancreatic islet cells of the neonatal rat is associated with a reduced expression of insulin-like growth factor II that may act as a survival factor. Endocrinology 139, 2994–3004 (1998).
Hill, D.J.. Increased and persistent circulating insulin-like growth factor II in neonatal transgenic mice suppresses developmental apoptosis in the pancreatic islets. Endocrinology 141, 1151–7 (2000).
Hogg, J., Hill, D.J. & Han, V.K. The ontogeny of insulin-like growth factor (IGF) and IGF-binding protein gene expression in the rat pancreas. J Mol Endocrinol 13, 49–58 (1994).
Petrik, J.. Overexpression of insulin-like growth factor-II in transgenic mice is associated with pancreatic islet cell hyperplasia. Endocrinology 140, 2353–63 (1999).
Reusens, B. & Remacle, C. Programming of the endocrine pancreas by the early nutritional environment. Int J Biochem Cell Biol 38, 913–22 (2006).
Montanya, E., Nacher, V., Biarnes, M. & Soler, J. Linear correlation between beta-cell mass and body weight throughout the lifespan in Lewis rats: role of beta-cell hyperplasia and hypertrophy. Diabetes 49, 1341–6 (2000).
Marynissen, G., Aerts, L. & Van Assche, F.A. The endocrine pancreas during pregnancy and lactation in the rat. J Dev Physiol 5, 373–81 (1983).
Van Assche, F.A., Aerts, L. & De Prins, F. A morphological study of the endocrine pancreas in human pregnancy. Br J Obstet Gynaecol 85, 818–20 (1978).
Scaglia, L., Smith, F.E. & Bonner-Weir, S. Apoptosis contributes to the involution of beta cell mass in the post partum rat pancreas. Endocrinology 136, 5461–8 (1995).
Chen, C., Hosokawa, H., Bumbalo, L.M. & Leahy, J.L. Mechanism of compensatory hyperinsulinemia in normoglycemic insulin-resistant spontaneously hypertensive rats. Augmented enzymatic activity of glucokinase in beta-cells. J Clin Invest 94, 399–404 (1994).
Liu, Y.Q., Jetton, T.L. & Leahy, J.L. Beta-cell adaptation to insulin resistance. Increased pyruvate carboxylase and malate-pyruvate shuttle activity in islets of nondiabetic Zucker fatty rats. J Biol Chem 277, 39163–8 (2002).
Butler, A.E.. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52, 102–10 (2003).
Jetton, T.L.. Mechanisms of compensatory beta-cell growth in insulin-resistant rats: roles of Akt kinase. Diabetes 54, 2294–304 (2005).
Kloppel, G., Lohr, M., Habich, K., Oberholzer, M. & Heitz, P.U. Islet pathology and the pathogenesis of type 1 and type 2 diabetes mellitus revisited. Surv Synth Pathol Res 4, 110–25 (1985).
Steil, G.M.. Adaptation of beta-cell mass to substrate oversupply: enhanced function with normal gene expression. Am J Physiol Endocrinol Metab 280, E788–96 (2001).
Biddinger, S.B. & Kahn, C.R. From mice to men: insights into the insulin resistance syndromes. Annu Rev Physiol 68, 123–58 (2006).
Rhodes, C.J. & White, M.F. Molecular insights into insulin action and secretion. Eur J Clin Invest 32 Suppl 3, 3–13 (2002).
Lupi, R.. 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 51, 1437–42 (2002).
Pick, A.. 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 47, 358–64 (1998).
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).
Danial, N.N.. Dual role of proapoptotic BAD in insulin secretion and beta cell survival. Nat Med 14, 144–53 (2008).
Liadis, N.. Distinct in vivo roles of caspase-8 in beta-cells in physiological and diabetes models. Diabetes 56, 2302–11 (2007).
Accili, D., Kido, Y., Nakae, J., Lauro, D. & Park, B.C. Genetics of type 2 diabetes: insight from targeted mouse mutants. Curr Mol Med 1, 9–23 (2001).
Bell, G.I. & Polonsky, K.S. Diabetes mellitus and genetically programmed defects in beta-cell function. Nature 414, 788–91 (2001).
Dickson, L.M. & Rhodes, C.J. Pancreatic beta-cell growth and survival in the onset of type 2 diabetes: a role for protein kinase B in the Akt? Am J Physiol Endocrinol Metab 287, E192–8 (2004).
Weir, G.C., Laybutt, D.R., Kaneto, H., Bonner-Weir, S. & Sharma, A. Beta-cell adaptation and decompensation during the progression of diabetes. Diabetes 50 Suppl 1, S154–9 (2001).
Donath, M.Y. & Halban, P.A. Decreased beta-cell mass in diabetes: significance, mechanisms and therapeutic implications. Diabetologia 47, 581–9 (2004).
Kahn, S.E. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 46, 3–19 (2003).
Mathis, D., Vence, L. & Benoist, C. Beta-cell death during progression to diabetes. Nature 414, 792–8 (2001).
Haataja, L., Gurlo, T., Huang, C.J. & Butler, P.C. Islet amyloid in type 2 diabetes, and the toxic oligomer hypothesis. Endocr Rev 29, 303–16 (2008).
Prentki, M. & Nolan, C.J. Islet beta cell failure in type 2 diabetes. J Clin Invest 116, 1802–12 (2006).
Rhodes, C.J. Type 2 diabetes-a matter of beta-cell life and death? Science 307, 380–4 (2005).
Donath, M.Y., Storling, J., Maedler, K. & Mandrup-Poulsen, T. Inflammatory mediators and islet beta-cell failure: a link between type 1 and type 2 diabetes. J Mol Med 81, 455–70 (2003).
Shoda, L.K.. A comprehensive review of interventions in the NOD mouse and implications for translation. Immunity 23, 115–26 (2005).
Finegood, D.T., Scaglia, L. & Bonner-Weir, S. Dynamics of beta-cell mass in the growing rat pancreas. Estimation with a simple mathematical model. Diabetes 44, 249–56 (1995).
Trudeau, J.D.. Neonatal beta-cell apoptosis: a trigger for autoimmune diabetes? Diabetes 49, 1–7 (2000).
Turley, S., Poirot, L., Hattori, M., Benoist, C. & Mathis, D. Physiological beta cell death triggers priming of self-reactive T cells by dendritic cells in a type-1 diabetes model. J Exp Med 198, 1527–37 (2003).
Faideau, B., Larger, E., Lepault, F., Carel, J.C. & Boitard, C. Role of beta-cells in type 1 diabetes pathogenesis. Diabetes 54 Suppl 2, S87–96 (2005).
Yoon, J.W., Jun, H.S. & Santamaria, P Cellular and molecular mechanisms for the initiation and progression of beta cell destruction resulting from the collaboration between macrophages and T cells. Autoimmunity 27, 109–22 (1998).
Bolitho, P., Voskoboinik, I., Trapani, J.A. & Smyth, M.J. Apoptosis induced by the lymphocyte effector molecule perforin. Current Opin Immunol 19, 339–47 (2007).
Cullen, S.P. & Martin, S.J. Mechanisms of granule-dependent killing. Cell Death Differ 15, 251–62 (2008).
Adeghate, E., Schattner, P. & Dunn, E. An update on the etiology and epidemiology of diabetes mellitus. Ann N Y Acad Sci 1084, 1–29 (2006).
Kim, M.S. & Polychronakos, C. Immunogenetics of type 1 diabetes. Hormone Res 64, 180–8 (2005).
Todd, J.A.. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 39, 857–64 (2007).
Van Der Werf, N., Kroese, F.G., Rozing, J. & Hillebrands, J.L. Viral infections as potential triggers of type 1 diabetes. Diabetes Metab Res Rev 23, 169–83 (2007).
Ylipaasto, P.. Global profiling of coxsackievirus- and cytokine-induced gene expression in human pancreatic islets. Diabetologia 48, 1510–22 (2005).
Harkonen, T., Lankinen, H., Davydova, B., Hovi, T. & Roivainen, M. Enterovirus infection can induce immune responses that cross-react with beta-cell autoantigen tyrosine phosphatase IA-2/IAR. J Med Virol 66, 340–50 (2002).
Jorns, A.. Immune cell infiltration, cytokine expression, and beta-cell apoptosis during the development of type 1 diabetes in the spontaneously diabetic LEW.1AR1/Ztm-iddm rat. Diabetes 54, 2041–52 (2005).
O’Brien, B.A., Harmon, B.V., Cameron, D.P. & Allan, D.J. Apoptosis is the mode of beta-cell death responsible for the development of IDDM in the nonobese diabetic (NOD) mouse. Diabetes 46, 750–7 (1997).
Pipeleers, D.. Role of pancreatic beta-cells in the process of beta-cell death. Diabetes 50 Suppl 1, S52–57 (2001).
Corbett, J.A., Wang, J.L., Sweetland, M.A., Lancaster, J.R., Jr. & McDaniel, M.L. Interleukin 1 beta induces the formation of nitric oxide by beta-cells purified from rodent islets of Langerhans. Evidence for the beta-cell as a source and site of action of nitric oxide. J Clin Invest 90, 2384–91 (1992).
Eizirik, D.L. & Mandrup-Poulsen, T. A choice of death–the signal-transduction of immune-mediated beta-cell apoptosis. Diabetologia 44, 2115–33 (2001).
Allison, J., Thomas, H.E., Catterall, T., Kay, T.W. & Strasser, A. Transgenic expression of dominant-negative Fas-associated death domain protein in beta cells protects against Fas ligand-induced apoptosis and reduces spontaneous diabetes in nonobese diabetic mice. J Immunol 175, 293–301 (2005).
Chervonsky, A.V.. The role of Fas in autoimmune diabetes. Cell 89, 17–24 (1997).
Dudek, N.L.. Cytotoxic T-cells from T-cell receptor transgenic NOD8.3 mice destroy beta-cells via the perforin and Fas pathways. Diabetes 55, 2412–8 (2006).
Itoh, N.. Requirement of Fas for the development of autoimmune diabetes in nonobese diabetic mice. J Exp Med 186, 613–18 (1997).
Kreuwel, H.T.. Comparing the relative role of perforin/granzyme versus Fas/Fas ligand cytotoxic pathways in CD8+ T cell-mediated insulin-dependent diabetes mellitus. J Immunol 163, 4335–41 (1999).
Thomas, H.E.. IL-1 receptor deficiency slows progression to diabetes in the NOD mouse. Diabetes 53, 113–21 (2004).
Vence, L., Benoist, C. & Mathis, D. Fas deficiency prevents type 1 diabetes by inducing hyporesponsiveness in islet beta-cell-reactive T-cells. Diabetes 53, 2797–803 (2004).
Danial, N.N. & Korsmeyer, S.J. Cell death: critical control points. Cell 116, 205–19 (2004).
Peter, M.E. & Krammer, P.H. The CD95(APO-1/Fas) DISC and beyond. Cell Death Differ 10, 26–35 (2003).
Aravind, L., Dixit, V.M. & Koonin, E.V. The domains of death: evolution of the apoptosis machinery. Trends Biochem Sci 24, 47–53 (1999).
Fesik, S.W. Insights into programmed cell death through structural biology. Cell 103, 273–82 (2000).
Boatright, K.M.. A unified model for apical caspase activation. Mol Cell 11, 529–41 (2003).
Donepudi, M., Mac Sweeney, A., Briand, C. & Grutter, M.G. Insights into the regulatory mechanism for caspase-8 activation. Mol Cell 11, 543–9 (2003).
McKenzie, M.D.. Proapoptotic BH3-only protein Bid is essential for death receptor-induced apoptosis of pancreatic beta-cells. Diabetes 57, 1284–92 (2008).
Scaffidi, C.. Differential modulation of apoptosis sensitivity in CD95 type I and type II cells. J Biol Chem 274, 22532–8 (1999).
Rabinovitch, A.. Transfection of human pancreatic islets with an anti-apoptotic gene (bcl-2) protects beta-cells from cytokine-induced destruction. Diabetes 48, 1223–9 (1999).
Sung, H.H.. Transgenic expression of decoy receptor 3 protects islets from spontaneous and chemical-induced autoimmune destruction in nonobese diabetic mice. J Exp Med 199, 1143–51 (2004).
Apostolou, I., Hao, Z., Rajewsky, K. & von Boehmer, H. Effective destruction of Fas-deficient insulin-producing beta cells in type 1 diabetes. J Exp Med 198, 1103–1106 (2003).
Cottet, S.. cFLIP protein prevents tumor necrosis factor-alpha-mediated induction of caspase-8-dependent apoptosis in insulin-secreting betaTc-Tet cells. Diabetes 51, 1805–14 (2002).
Irmler, M.. Inhibition of death receptor signals by cellular FLIP. Nature 388, 190–5 (1997).
Maedler, K.. FLIP switches Fas-mediated glucose signaling in human pancreatic beta cells from apoptosis to cell replication. Proc Natl Acad Sci U S A 99, 8236–41 (2002).
Millet, I.. Targeted expression of the anti-apoptotic gene CrmA to NOD pancreatic islets protects from autoimmune diabetes. J Autoimmun 26, 7–15 (2006).
Schumann, D.M.. The Fas pathway is involved in pancreatic beta cell secretory function. Proc Natl Acad Sci U S A 104, 2861–6 (2007).
Park, S.M., Schickel, R. & Peter, M.E. Nonapoptotic functions of FADD-binding death receptors and their signaling molecules. Curr Opin Cell Biol 17, 610–16 (2005).
Micheau, O. & Tschopp, J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 114, 181–90 (2003).
Chen, G. & Goeddel, D.V. TNF-R1 signaling: a beautiful pathway. Science 296, 1634–5 (2002).
Muppidi, J.R., Tschopp, J. & Siegel, R.M. Life and death decisions: secondary complexes and lipid rafts in TNF receptor family signal transduction. Immunity 21, 461–5 (2004).
Chu, Z.L.. Suppression of tumor necrosis factor-induced cell death by inhibitor of apoptosis c-IAP2 is under NF-kappaB control. Proc Natl Acad Sci U S A 94, 10057–62 (1997).
Wang, C.Y., Mayo, M.W., Korneluk, R.G., Goeddel, D.V. & Baldwin, A.S., Jr. NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 281, 1680–3 (1998).
Kreuz, S., Siegmund, D., Scheurich, P. & Wajant, H. NF-kappaB inducers upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor signaling. Mol Cell Biol 21, 3964–73 (2001).
Micheau, O., Lens, S., Gaide, O., Alevizopoulos, K. & Tschopp, J. NF-kappaB signals induce the expression of c-FLIP. Mol Cell Biol 21, 5299–305 (2001).
Melloul, D. Role of NF-kappaB in beta-cell death. Biochem Soc Trans 36, 334–9 (2008).
Hansen, B.S.. Effect of interleukin-1 on the biosynthesis of proinsulin and insulin in isolated rat pancreatic islets. Biomed Biochim Acta 47, 305–9 (1988).
Maedler, K.. Low concentration of interleukin-1beta induces FLICE-inhibitory protein-mediated beta-cell proliferation in human pancreatic islets. Diabetes 55, 2713–22 (2006).
Maedler, K.. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest 110, 851–60 (2002).
Sparre, T.. IL-1beta induced protein changes in diabetes prone BB rat islets of Langerhans identified by proteome analysis. Diabetologia 45, 1550–61 (2002).
Arnush, M.. IL-1 produced and released endogenously within human islets inhibits beta cell function. J Clin Invest 102, 516–26 (1998).
Spinas, G.A.. The bimodal effect of interleukin 1 on rat pancreatic beta-cells – stimulation followed by inhibition – depends upon dose, duration of exposure, and ambient glucose concentration. Acta Endocrinol 119, 307–11 (1988).
Dupraz, P.. Dominant negative MyD88 proteins inhibit interleukin-1beta/interferon-gamma-mediated induction of nuclear factor kappa B-dependent nitrite production and apoptosis in beta cells. J Biol Chem 275, 37672–8 (2000).
Tellez, N.. Adenoviral overexpression of interleukin-1 receptor antagonist protein increases beta-cell replication in rat pancreatic islets. Gene Ther 12, 120–8 (2005).
Dinarello, C.A. Interleukin-1. Cytokine Growth Factor Rev 8, 253–65 (1997).
Burns, K.. MyD88, an adapter protein involved in interleukin-1 signaling. J Biol Chem 273, 12203–9 (1998).
Cao, Z., Xiong, J., Takeuchi, M., Kurama, T. & Goeddel, D.V. TRAF6 is a signal transducer for interleukin-1. Nature 383, 443–6 (1996).
Verstrepen, L.. TLR-4, IL-1R and TNF-R signaling to NF-kappaB: variations on a common theme. Cell Mol Life Sci 65, 2964–78 (2008).
Cardozo, A.K.. A comprehensive analysis of cytokine-induced and nuclear factor-kappa B-dependent genes in primary rat pancreatic beta-cells. J Biol Chem 276, 48879–86 (2001).
Eldor, R.. Conditional and specific NF-kappaB blockade protects pancreatic beta cells from diabetogenic agents. Proc Natl Acad Sci U S A 103, 5072–7 (2006).
Suk, K.. IFN-gamma/TNF-alpha synergism as the final effector in autoimmune diabetes: a key role for STAT1/IFN regulatory factor-1 pathway in pancreatic beta cell death. J Immunol 166, 4481–9 (2001).
O’Shea, J.J. & Murray, P.J. Cytokine signaling modules in inflammatory responses. Immunity 28, 477–87 (2008).
Krebs, D.L. & Hilton, D.J. SOCS proteins: negative regulators of cytokine signaling. Stem Cells (Dayton, Ohio) 19, 378–87 (2001).
Qing, Y., Costa-Pereira, A.P., Watling, D. & Stark, G.R. Role of tyrosine 441 of interferon-gamma receptor subunit 1 in SOCS-1-mediated attenuation of STAT1 activation. J Biol Chem 280, 1849–53 (2005).
Gysemans, C.A.. Disruption of the gamma-interferon signaling pathway at the level of signal transducer and activator of transcription-1 prevents immune destruction of beta-cells. Diabetes 54, 2396–403 (2005).
Chong, M.M., Thomas, H.E. & Kay, T.W. Gamma-interferon signaling in pancreatic beta-cells is persistent but can be terminated by overexpression of suppressor of cytokine signaling-1. Diabetes 50, 2744–51 (2001).
Chong, M.M., Thomas, H.E. & Kay, T.W. Suppressor of cytokine signaling-1 regulates the sensitivity of pancreatic beta cells to tumor necrosis factor. J Biol Chem 277, 27945–52 (2002).
Flodstrom-Tullberg, M.. Target cell expression of suppressor of cytokine signaling-1 prevents diabetes in the NOD mouse. Diabetes 52, 2696–700 (2003).
Karlsen, A.E.. Suppressor of cytokine signaling 3 (SOCS-3) protects beta-cells against interleukin-1beta– and interferon-gamma–mediated toxicity. Proc Natl Acad Sci U S A 98, 12191–6 (2001).
Solomon, M., Flodstrom-Tullberg, M. & Sarvetnick, N. Differences in suppressor of cytokine signaling-1 (SOCS-1) expressing islet allograft destruction in normal BALB/c and spontaneously-diabetic NOD recipient mice. Transplantation 79, 1104–9 (2005).
Green, I.C.. Effects of cytokines and nitric oxide donors on insulin secretion, cyclic GMP and DNA damage: relation to nitric oxide production. Biochem Soc Trans 22, 30–7 (1994).
Riboulet-Chavey, A., Diraison, F., Siew, L.K., Wong, F.S. & Rutter, G.A. Inhibition of AMP-activated protein kinase protects pancreatic beta-cells from cytokine-mediated apoptosis and CD8+ T-cell-induced cytotoxicity. Diabetes 57, 415–23 (2008).
Bolaffi, J.L., Rodd, G.G., Wang, J. & Grodsky, G.M. Interrelationship of changes in islet nicotine adeninedinucleotide, insulin secretion, and cell viability induced by interleukin-1 beta. Endocrinology 134, 537–42 (1994).
Iqbal, J. & Zaidi, M. TNF regulates cellular NAD+ metabolism in primary macrophages. Biochem Biophys Res Commun 342, 1312–18 (2006).
Radons, J.. Nitric oxide toxicity in islet cells involves poly(ADP-ribose) polymerase activation and concomitant NAD+ depletion. Biochem Biophys Res Commun 199, 1270–7 (1994).
Chang, I.. Role of calcium in pancreatic islet cell death by IFN-gamma/TNF-alpha. J Immunol 172, 7008–14 (2004).
Grankvist, K., Marklund, S.L. & Taljedal, I.B. CuZn-superoxide dismutase, Mn-superoxide dismutase, catalase and glutathione peroxidase in pancreatic islets and other tissues in the mouse. Biochem J 199, 393–8 (1981).
Tiedge, M., Lortz, S., Drinkgern, J. & Lenzen, S. Relation between antioxidant enzyme gene expression and antioxidative defense status of insulin-producing cells. Diabetes 46, 1733–1742 (1997).
Welsh, N.. Differences in the expression of heat-shock proteins and antioxidant enzymes between human and rodent pancreatic islets: implications for the pathogenesis of insulin-dependent diabetes mellitus. Mol Med 1, 806–20 (1995).
Cahuana, G.M.. Nitric oxide mediates the survival action of IGF-1 and insulin in pancreatic beta cells. Cell Signal 20, 301–10 (2008).
Kitiphongspattana, K.. Protective role for nitric oxide during the endoplasmic reticulum stress response in pancreatic beta-cells. Am J Physiol Endocrinol Metab 292, E1543–54 (2007).
Hohmeier, H.E., Thigpen, A., Tran, V.V., Davis, R. & Newgard, C.B. Stable expression of manganese superoxide dismutase (MnSOD) in insulinoma cells prevents IL-1beta- induced cytotoxicity and reduces nitric oxide production. J Clin Invest 101, 1811–20 (1998).
Lortz, S.. Protection of insulin-producing RINm5F cells against cytokine-mediated toxicity through overexpression of antioxidant enzymes. Diabetes 49, 1123–30 (2000).
Flodstrom, M., Tyrberg, B., Eizirik, D.L. & Sandler, S. Reduced sensitivity of inducible nitric oxide synthase-deficient mice to multiple low-dose streptozotocin-induced diabetes. Diabetes 48, 706–713 (1999).
Hotta, M.. Pancreatic beta cell-specific expression of thioredoxin, an antioxidative and antiapoptotic protein, prevents autoimmune and streptozotocin-induced diabetes. J Exp Med 188, 1445–51 (1998).
Kim, J.Y.. Inhibition of diabetes in non-obese diabetic mice by nicotinamide treatment for 5 weeks at the early age. J Korean Med Sci 12, 293–7 (1997).
Kubisch, H.M.. Transgenic copper/zinc superoxide dismutase modulates susceptibility to type I diabetes. Proc Natl Acad Sci U S A 91, 9956–9 (1994).
O’Brien, B.A., Harmon, B.V., Cameron, D.P. & Allan, D.J. Nicotinamide prevents the development of diabetes in the cyclophosphamide-induced NOD mouse model by reducing beta-cell apoptosis. J Pathol 191, 86–92 (2000).
Piganelli, J.D.. A metalloporphyrin-based superoxide dismutase mimic inhibits adoptive transfer of autoimmune diabetes by a diabetogenic T-cell clone. Diabetes 51, 347–55 (2002).
Reddy, S., Karanam, M. & Robinson, E. Prevention of cyclophosphamide-induced accelerated diabetes in the NOD mouse by nicotinamide or a soy protein-based infant formula. Int J Exp Diabetes Res 1, 299–313 (2001).
Suarez-Pinzon, W.L., Mabley, J.G., Power, R., Szabo, C. & Rabinovitch, A. Poly (ADP-ribose) polymerase inhibition prevents spontaneous and recurrent autoimmune diabetes in NOD mice by inducing apoptosis of islet-infiltrating leukocytes. Diabetes 52, 1683–8 (2003).
Yamada, K.. Preventive and therapeutic effects of large-dose nicotinamide injections on diabetes associated with insulitis. An observation in nonobese diabetic (NOD) mice. Diabetes 31, 749–53 (1982).
Liu, D.. Cytokines induce apoptosis in beta-cells isolated from mice lacking the inducible isoform of nitric oxide synthase (iNOS-/-). Diabetes 49, 1116–22 (2000).
Suarez-Pinzon, W.L.. An inhibitor of inducible nitric oxide synthase and scavenger of peroxynitrite prevents diabetes development in NOD mice. J Autoimmun 16, 449–55 (2001).
Szabo, C. Roles of poly(ADP-ribose) polymerase activation in the pathogenesis of diabetes mellitus and its complications. Pharmacol Res 52, 60–71 (2005).
Lenzen, S. The mechanisms of alloxan- and streptozotocin-induced diabetes. Diabetologia 51, 216–26 (2008).
Burkart, V.. Mice lacking the poly(ADP-ribose) polymerase gene are resistant to pancreatic beta-cell destruction and diabetes development induced by streptozocin. Nat Med 5, 314–19 (1999).
Masutani, M.. Poly(ADP-ribose) polymerase gene disruption conferred mice resistant to streptozotocin-induced diabetes. Proc Natl Acad Sci U S A 96, 2301–4 (1999).
Pieper, A.A.. Poly(ADP-ribose) polymerase-deficient mice are protected from streptozotocin-induced diabetes. Proc Natl Acad Sci U S A 96, 3059–64 (1999).
Papaccio, G.. Interleukin (IL)-1beta toxicity to islet beta cells: efaroxan exerts a complete protection. J Cell Physiol 203, 94–102 (2005).
Tortora, V., Quijano, C., Freeman, B., Radi, R. & Castro, L. Mitochondrial aconitase reaction with nitric oxide, S-nitrosoglutathione, and peroxynitrite: mechanisms and relative contributions to aconitase inactivation. Free Radic Biol Med 42, 1075–88 (2007).
Wilson, G.L., Patton, N.J. & LeDoux, S.P. Mitochondrial DNA in beta-cells is a sensitive target for damage by nitric oxide. Diabetes 46, 1291–5 (1997).
Leahy, J.L. Pathogenesis of type 2 diabetes mellitus. Arch Med Res 36, 197–209 (2005).
Martin, B.C.. Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet 340, 925–9 (1992).
Muoio, D.M. & Newgard, C.B. Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat Rev 9, 193–205 (2008).
Godsland, I.F., Jeffs, J.A. & Johnston, D.G. Loss of beta cell function as fasting glucose increases in the non-diabetic range. Diabetologia 47, 1157–66 (2004).
Nolan, C.J.. Beta cell compensation for insulin resistance in Zucker fatty rats: increased lipolysis and fatty acid signalling. Diabetologia 49, 2120–30 (2006).
Poitout, V. & Robertson, R.P. Glucolipotoxicity: fuel excess and beta-cell dysfunction. Endocr Rev 29, 351–66 (2008).
El-Assaad, W.. Saturated fatty acids synergize with elevated glucose to cause pancreatic beta-cell death. Endocrinology 144, 4154–63 (2003).
Maedler, K., Oberholzer, J., Bucher, P., Spinas, G.A. & Donath, M.Y. Monounsaturated fatty acids prevent the deleterious effects of palmitate and high glucose on human pancreatic beta-cell turnover and function. Diabetes 52, 726–33 (2003).
Maedler, K.. Distinct effects of saturated and monounsaturated fatty acids on beta-cell turnover and function. Diabetes 50, 69–76 (2001).
Maestre, I.. Mitochondrial dysfunction is involved in apoptosis induced by serum withdrawal and fatty acids in the beta-cell line INS-1. Endocrinology 144, 335–45 (2003).
Shimabukuro, M., Wang, M.Y., Zhou, Y.T., Newgard, C.B. & Unger, R.H. Protection against lipoapoptosis of beta cells through leptin-dependent maintenance of Bcl-2 expression. Proc Natl Acad Sci U S A 95, 9558–61 (1998).
Prentki, M. & Corkey, B.E. Are the beta-cell signaling molecules malonyl-CoA and cystolic long-chain acyl-CoA implicated in multiple tissue defects of obesity and NIDDM? Diabetes 45, 273–83 (1996).
Cline, G.W., Lepine, R.L., Papas, K.K., Kibbey, R.G. & Shulman, G.I. 13C NMR isotopomer analysis of anaplerotic pathways in INS-1 cells. J Biol Chem 279, 44370–5 (2004).
Farfari, S., Schulz, V., Corkey, B. & Prentki, M. Glucose-regulated anaplerosis and cataplerosis in pancreatic beta-cells: possible implication of a pyruvate/citrate shuttle in insulin secretion. Diabetes 49, 718–26 (2000).
McGarry, J.D. & Brown, N.F. The mitochondrial carnitine palmitoyltransferase system. From concept to molecular analysis. Eur J Biochem 244, 1–14 (1997).
Roduit, R.. A role for the malonyl-CoA/long-chain acyl-CoA pathway of lipid signaling in the regulation of insulin secretion in response to both fuel and nonfuel stimuli. Diabetes 53, 1007–19 (2004).
Salt, I.P., Johnson, G., Ashcroft, S.J. & Hardie, D.G. AMP-activated protein kinase is activated by low glucose in cell lines derived from pancreatic beta cells, and may regulate insulin release. Biochem J 335 (Pt 3), 533–9 (1998).
Paumen, M.B., Ishida, Y., Muramatsu, M., Yamamoto, M. & Honjo, T. Inhibition of carnitine palmitoyltransferase I augments sphingolipid synthesis and palmitate-induced apoptosis. J Biol Chem 272, 3324–9 (1997).
Revoltella, R.P., Dal Canto, B., Caracciolo, L. & D’Urso, C.M. L-carnitine and some of its analogs delay the onset of apoptotic cell death initiated in murine C2.8 hepatocytic cells after hepatocyte growth factor deprivation. Biochim Biophys Acta 1224, 333–41 (1994).
Ruderman, N. & Prentki, M. AMP kinase and malonyl-CoA: targets for therapy of the metabolic syndrome. Nat Rev Drug Discov 3, 340–51 (2004).
Cnop, M., Hannaert, J.C., Hoorens, A., Eizirik, D.L. & Pipeleers, D.G. Inverse relationship between cytotoxicity of free fatty acids in pancreatic islet cells and cellular triglyceride accumulation. Diabetes 50, 1771–7 (2001).
Hardy, S.. Saturated fatty acid-induced apoptosis in MDA-MB-231 breast cancer cells. A role for cardiolipin. J Biol Chem 278, 31861–70 (2003).
Tuominen, E.K., Wallace, C.J. & Kinnunen, P.K. Phospholipid-cytochrome c interaction: evidence for the extended lipid anchorage. J Biol Chem 277, 8822–6 (2002).
Zhang, M., Mileykovskaya, E. & Dowhan, W. Gluing the respiratory chain together. Cardiolipin is required for supercomplex formation in the inner mitochondrial membrane. J Biol Chem 277, 43553–6 (2002).
Carlsson, C., Borg, L.A. & Welsh, N. Sodium palmitate induces partial mitochondrial uncoupling and reactive oxygen species in rat pancreatic islets in vitro. Endocrinology 140, 3422–8 (1999).
Ott, M., Zhivotovsky, B. & Orrenius, S. Role of cardiolipin in cytochrome c release from mitochondria. Cell Death Differ 14, 1243–7 (2007).
Buratta, M.. Loss of cardiolipin in palmitate-treated GL15 glioblastoma cells favors cytochrome c release from mitochondria leading to apoptosis. J Neurochem 105, 1019–31 (2008).
Ostrander, D.B., Sparagna, G.C., Amoscato, A.A., McMillin, J.B. & Dowhan, W. Decreased cardiolipin synthesis corresponds with cytochrome c release in palmitate-induced cardiomyocyte apoptosis. J Biol Chem 276, 38061–7 (2001).
Shimabukuro, M.. Lipoapoptosis in beta-cells of obese prediabetic fa/fa rats. Role of serine palmitoyltransferase overexpression. J Biol Chem 273, 32487–90 (1998).
Paumen, M.B.. Direct interaction of the mitochondrial membrane protein carnitine palmitoyltransferase I with Bcl-2. Biochem Biophys Res Commun 231, 523–5 (1997).
Kashkar, H., Wiegmann, K., Yazdanpanah, B., Haubert, D. & Kronke, M. Acid sphingomyelinase is indispensable for UV light-induced Bax conformational change at the mitochondrial membrane. J Biol Chem 280, 20804–13 (2005).
Mercie, P.. Comparative methodologic study of NFkappaB activation in cultured endothelial cells. J Lab Clin Med 136, 402–11 (2000).
Heinrich, M.. Cathepsin D links TNF-induced acid sphingomyelinase to Bid-mediated caspase-9 and -3 activation. Cell Death Differ 11, 550–63 (2004).
Hardy, S., Langelier, Y. & Prentki, M. Oleate activates phosphatidylinositol 3-kinase and promotes proliferation and reduces apoptosis of MDA-MB-231 breast cancer cells, whereas palmitate has opposite effects. Cancer Res 60, 6353–8 (2000).
Zundel, W. & Giaccia, A. Inhibition of the anti-apoptotic PI(3)K/Akt/Bad pathway by stress. Genes Dev 12, 1941–6 (1998).
Basu, S., Bayoumy, S., Zhang, Y., Lozano, J. & Kolesnick, R. BAD enables ceramide to signal apoptosis via Ras and Raf-1. J Biol Chem 273, 30419–26 (1998).
Bandyopadhyay, S.. Mechanism of apoptosis induced by the inhibition of fatty acid synthase in breast cancer cells. Cancer Res 66, 593440 (2006).
Corkey, B.E.. A role for malonyl-CoA in glucose-stimulated insulin secretion from clonal pancreatic beta-cells. J Biol Chem 264, 21608–12 (1989).
Prentki, M.. Malonyl-CoA and long chain acyl-CoA esters as metabolic coupling factors in nutrient-induced insulin secretion. J Biol Chem 267, 5802–10 (1992).
Cardozo, A.K.. Cytokines downregulate the sarcoendoplasmic reticulum pump Ca2+ ATPase 2b and deplete endoplasmic reticulum Ca2+, leading to induction of endoplasmic reticulum stress in pancreatic beta-cells. Diabetes 54, 452–61 (2005).
Zhou, Y.P.. Apoptosis in insulin-secreting cells. Evidence for the role of intracellular Ca2+ stores and arachidonic acid metabolism. J Clin Invest 101, 1623–32 (1998).
Scheuner, D. & Kaufman, R.J. The unfolded protein response: a pathway that links insulin demand with beta-cell failure and diabetes. Endocr Rev 29, 317–33 (2008).
Sanke, T., Bell, G.I., Sample, C., Rubenstein, A.H. & Steiner, D.F. An islet amyloid peptide is derived from an 89-amino acid precursor by proteolytic processing. J Biol Chem 263, 17243–6 (1988).
Arnelo, U.. Chronic low dose islet amyloid polypeptide infusion reduces food intake, but does not influence glucose metabolism, in unrestrained conscious rats: studies using a novel aortic catheterization technique. Endocrinology 138, 4081–5 (1997).
Ahren, B., Oosterwijk, C., Lips, C.J. & Hoppener, J.W. Transgenic overexpression of human islet amyloid polypeptide inhibits insulin secretion and glucose elimination after gastric glucose gavage in mice. Diabetologia 41, 1374–80 (1998).
Tokuyama, T.. Expression of human islet amyloid polypeptide/amylin impairs insulin secretion in mouse pancreatic beta cells. Metab Clin Exp 46, 1044–51 (1997).
Meier, J.J.. Inhibition of human IAPP fibril formation does not prevent beta-cell death: evidence for distinct actions of oligomers and fibrils of human IAPP. Am J Physiol Endocrinol Metab 291, E1317–24 (2006).
Bretherton-Watt, D.. Altered islet amyloid polypeptide (amylin) gene expression in rat models of diabetes. Diabetologia 32, 881–3 (1989).
Janson, J.. Spontaneous diabetes mellitus in transgenic mice expressing human islet amyloid polypeptide. Proc Natl Acad Sci U S A 93, 7283–8 (1996).
Kahn, S.E., Andrikopoulos, S. & Verchere, C.B. Islet amyloid: a long-recognized but underappreciated pathological feature of type 2 diabetes. Diabetes 48, 241–53 (1999).
Lorenzo, A., Razzaboni, B., Weir, G.C. & Yankner, B.A. Pancreatic islet cell toxicity of amylin associated with type-2 diabetes mellitus. Nature 368, 756–60 (1994).
Kayed, R.. Fibril specific, conformation dependent antibodies recognize a generic epitope common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar oligomers. Mol Neurodegener 2, 18 (2007).
Kayed, R.. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 300, 486–9 (2003).
Glabe, C.G. Common mechanisms of amyloid oligomer pathogenesis in degenerative disease. Neurobiol Aging 27, 570–5 (2006).
Lindholm, D., Wootz, H. & Korhonen, L. ER stress and neurodegenerative diseases. Cell Death Differ 13, 385–92 (2006).
Butler, A.E.. Diabetes due to a progressive defect in beta-cell mass in rats transgenic for human islet amyloid polypeptide (HIP Rat): a new model for type 2 diabetes. Diabetes 53, 1509–16 (2004).
Couce, M.. Treatment with growth hormone and dexamethasone in mice transgenic for human islet amyloid polypeptide causes islet amyloidosis and beta-cell dysfunction. Diabetes 45, 1094–101 (1996).
Cooper, G.J.. Purification and characterization of a peptide from amyloid-rich pancreases of type 2 diabetic patients. Proc Natl Acad Sci U S A 84, 8628–32 (1987).
Westermark, P., Wernstedt, C., O’Brien, T.D., Hayden, D.W. & Johnson, K.H. Islet amyloid in type 2 human diabetes mellitus and adult diabetic cats contains a novel putative polypeptide hormone. Am J Pathol 127, 414–17 (1987).
Huang, C.J.. 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 56, 2016–27 (2007).
Laybutt, D.R.. Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 50, 752–63 (2007).
Marchetti, P.. The endoplasmic reticulum in pancreatic beta cells of type 2 diabetes patients. Diabetologia 50, 2486–94 (2007).
Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev 8, 519–29 (2007).
Fujita, E.. Two endoplasmic reticulum-associated degradation (ERAD) systems for the novel variant of the mutant dysferlin: ubiquitin/proteasome ERAD(I) and autophagy/lysosome ERAD(II). Hum Mol Genet 16, 618–29 (2007).
Berridge, M.J. The endoplasmic reticulum: a multifunctional signaling organelle. Cell Calcium 32, 235–49 (2002).
Hoyer-Hansen, M.. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol Cell 25, 193–205 (2007).
Szegezdi, E., Logue, S.E., Gorman, A.M. & Samali, A. Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep 7, 880–5 (2006).
Harding, H.P.. Diabetes mellitus and exocrine pancreatic dysfunction in perk-/- mice reveals a role for translational control in secretory cell survival. Mol Cell 7, 1153–63 (2001).
Zhang, P.. The PERK eukaryotic initiation factor 2 alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Mol Cell Biol 22, 3864–74 (2002).
Zhang, W.. PERK EIF2AK3 control of pancreatic beta cell differentiation and proliferation is required for postnatal glucose homeostasis. Cell metabolism 4, 491–7 (2006).
Scheuner, D.. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 7, 1165–76 (2001).
Allotey, R.A.. The EIF2AK3 gene region and type I diabetes in subjects from South India. Genes Immun 5, 648–52 (2004).
Castelnau, P.. Wolcott-Rallison syndrome: a case with endocrine and exocrine pancreatic deficiency and pancreatic hypotrophy. Eur J Pediatr 159, 631–633 (2000).
Delepine, M.. EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat Genet 25, 406–9 (2000).
Thornton, C.M., Carson, D.J. & Stewart, F.J. Autopsy findings in the Wolcott-Rallison syndrome. Pediatr Pathol Lab Med 17, 487–96 (1997).
Meex, S.J.. Activating transcription factor 6 polymorphisms and haplotypes are associated with impaired glucose homeostasis and type 2 diabetes in Dutch Caucasians. J Clin Endocrinol Metab 92, 2720–5 (2007).
Thameem, F., Farook, V.S., 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 55, 839–42 (2006).
Seino, S. S20G mutation of the amylin gene is associated with Type II diabetes in Japanese. Study group of comprehensive analysis of genetic factors in diabetes mellitus. Diabetologia 44, 906–9 (2001).
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, 1249–59 (2001).
Puthalakath, H.. ER stress triggers apoptosis by activating BH3-only protein Bim. Cell 129, 1337–49 (2007).
Futami, T., Miyagishi, M. & Taira, K. Identification of a network involved in thapsigargin-induced apoptosis using a library of small interfering RNA expression vectors. J Biol Chem 280, 826–31 (2005).
Li, J., Lee, B. & Lee, A.S. Endoplasmic reticulum stress-induced apoptosis: multiple pathways and activation of p53-up-regulated modulator of apoptosis (PUMA) and NOXA by p53. J Biol Chem 281, 7260–70 (2006).
Yamaguchi, H. & Wang, H.G. CHOP is involved in endoplasmic reticulum stress-induced apoptosis by enhancing DR5 expression in human carcinoma cells. J Biol Chem 279, 45495–502 (2004).
Zhang, S., Liu, H., Yu, H. & Cooper, G.J. Fas-associated death receptor signaling evoked by human amylin in islet beta-cells. Diabetes 57, 348–56 (2008).
Ohoka, N., Yoshii, S., Hattori, T., Onozaki, K. & Hayashi, H. TRB3, a novel ER stress-inducible gene, is induced via ATF4-CHOP pathway and is involved in cell death. EMBO J 24, 1243–55 (2005).
Oyadomari, S.. Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes. J Clin Invest 109, 525–32 (2002).
Oyadomari, S.. Nitric oxide-induced apoptosis in pancreatic beta cells is mediated by the endoplasmic reticulum stress pathway. Proc Natl Acad Sci U S A 98, 10845–50 (2001).
Huang, C.J.. Induction of endoplasmic reticulum stress-induced beta-cell apoptosis and accumulation of polyubiquitinated proteins by human islet amyloid polypeptide. Am J Physiol Endocrinol Metab 293, E1656–62 (2007).
Urano, F.. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287, 664–6 (2000).
Nishitoh, H.. ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev 16, 1345–55 (2002).
Bassik, M.C., Scorrano, L., Oakes, S.A., Pozzan, T. & Korsmeyer, S.J. Phosphorylation of BCL-2 regulates ER Ca2+ homeostasis and apoptosis. EMBO J 23, 1207–16 (2004).
Rong, Y. & Distelhorst, C.W. Bcl-2 protein family members: versatile regulators of calcium signaling in cell survival and apoptosis. Annu Rev Physiol 70, 73–91 (2008).
Hetz, C.. Bax channel inhibitors prevent mitochondrion-mediated apoptosis and protect neurons in a model of global brain ischemia. J Biol Chem 280, 42960–70 (2005).
Halban, P.A. Cellular sources of new pancreatic beta cells and therapeutic implications for regenerative medicine. Nat Cell Biol 6, 1021–25 (2004).
Limbert, C., Path, G., Jakob, F. & Seufert, J. Beta-cell replacement and regeneration: Strategies of cell-based therapy for type 1 diabetes mellitus. Diabetes Res Clin Pract 79, 389–99 (2008).
Carlsson, P.O., Palm, F., Andersson, A. & Liss, P. Markedly decreased oxygen tension in transplanted rat pancreatic islets irrespective of the implantation site. Diabetes 50, 489–495 (2001).
Carlsson, P.O., Palm, F. & Mattsson, G. Low revascularization of experimentally transplanted human pancreatic islets. J Clin Endocrinol Metab 87, 5418–23 (2002).
He, H., Stone, J.R. & Perkins, D.L. Analysis of robust innate immune response after transplantation in the absence of adaptive immunity. Transplantation 73, 853–61 (2002).
Mandrup-Poulsen, T. beta-cell apoptosis: stimuli and signaling. Diabetes 50 Suppl 1, S58–63 (2001).
Biarnes, M.. Beta-cell death and mass in syngeneically transplanted islets exposed to short- and long-term hyperglycemia. Diabetes 51, 66–72 (2002).
Davalli, A.M.. Vulnerability of islets in the immediate posttransplantation period. Dynamic changes in structure and function. Diabetes 45, 1161–7 (1996).
Ryan, E.A.. Successful islet transplantation: continued insulin reserve provides long-term glycemic control. Diabetes 51, 2148–57 (2002).
Emamaullee, J.A. & Shapiro, A.M. Interventional strategies to prevent beta-cell apoptosis in islet transplantation. Diabetes 55, 1907–14 (2006).
Mysore, T.B.. Overexpression of glutathione peroxidase with two isoforms of superoxide dismutase protects mouse islets from oxidative injury and improves islet graft function. Diabetes 54, 2109–16 (2005).
Gysemans, C.. Prevention of primary non-function of islet xenografts in autoimmune diabetic NOD mice by anti-inflammatory agents. Diabetologia 46, 1115–23 (2003).
Sandberg, J.O., Eizirik, D.L., Sandler, S., Tracey, D.E. & Andersson, A. Treatment with an interleukin-1 receptor antagonist protein prolongs mouse islet allograft survival. Diabetes 42, 1845–51 (1993).
Tellez, N.. Adenoviral overproduction of interleukin-1 receptor antagonist increases beta cell replication and mass in syngeneically transplanted islets, and improves metabolic outcome. Diabetologia 50, 602–11 (2007).
Ronn, S.G.. Suppressor of cytokine signalling-3 expression inhibits cytokine-mediated destruction of primary mouse and rat pancreatic islets and delays allograft rejection. Diabetologia 51, 1873–82 (2008).
Emamaullee, J., Liston, P., Korneluk, R.G., Shapiro, A.M. & Elliott, J.F. XIAP overexpression in islet beta-cells enhances engraftment and minimizes hypoxia-reperfusion injury. Am J Transplant 5, 1297–305 (2005).
Emamaullee, J.A.. XIAP overexpression in human islets prevents early posttransplant apoptosis and reduces the islet mass needed to treat diabetes. Diabetes 54, 2541–8 (2005).
Hui, H.. Adenovirus-mediated XIAP gene transfer reverses the negative effects of immunosuppressive drugs on insulin secretion and cell viability of isolated human islets. Diabetes 54, 424–33 (2005).
Plesner, A., Liston, P., Tan, R., Korneluk, R.G. & Verchere, C.B. The X-linked inhibitor of apoptosis protein enhances survival of murine islet allografts. Diabetes 54, 2533–40 (2005).
Dohi, T.. Inhibition of apoptosis by survivin improves transplantation of pancreatic islets for treatment of diabetes in mice. EMBO Rep 7, 438–43 (2006).
Montolio, M., Tellez, N., Biarnes, M., Soler, J. & Montanya, E. Short-term culture with the caspase inhibitor z-VAD.fmk reduces beta cell apoptosis in transplanted islets and improves the metabolic outcome of the graft. Cell Transplant 14, 59–65 (2005).
Dupraz, P.. Lentivirus-mediated Bcl-2 expression in betaTC-tet cells improves resistance to hypoxia and cytokine-induced apoptosis while preserving in vitro and in vivo control of insulin secretion. Gene Ther 6, 1160–9 (1999).
Klein, D.. Delivery of Bcl-XL or its BH4 domain by protein transduction inhibits apoptosis in human islets. Biochem Biophys Res Commun 323, 473–8 (2004).
Contreras, J.L.. Cytoprotection of pancreatic islets before and soon after transplantation by gene transfer of the anti-apoptotic Bcl-2 gene. Transplantation 71, 1015–23 (2001).
Pinton, P. & Rizzuto, R. Bcl-2 and Ca2+ homeostasis in the endoplasmic reticulum. Cell Death Differ 13, 1409–18 (2006).
Zhou, Y.P.. Overexpression of Bcl-x(L) in beta-cells prevents cell death but impairs mitochondrial signal for insulin secretion. Am J Physiol Endocrinol Metab 278, E340–51 (2000).
Danial, N.N.. BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis. Nature 424, 952–6 (2003).
Terauchi, Y.. Glucokinase and IRS-2 are required for compensatory beta cell hyperplasia in response to high-fat diet-induced insulin resistance. J Clin Invest 117, 246–57 (2007).
Bose, A.K., Mocanu, M.M., Carr, R.D., Brand, C.L. & Yellon, D.M. Glucagon-like peptide 1 can directly protect the heart against ischemia/reperfusion injury. Diabetes 54, 146–51 (2005).
Baggio, L.L. & Drucker, D.J. Biology of incretins: GLP-1 and GIP. Gastroenterology 132, 2131–57 (2007).
De Leon, D.D., Crutchlow, M.F., Ham, J.Y. & Stoffers, D.A. Role of glucagon-like peptide-1 in the pathogenesis and treatment of diabetes mellitus. Int J Biochem Cell Biol 38, 845–59 (2006).
Froud, T.. The use of exenatide in islet transplant recipients with chronic allograft dysfunction: safety, efficacy, and metabolic effects. Transplantation 86, 36–45 (2008).
King, P.J. The hypothalamus and obesity. Current Drug Targets 6, 225–40 (2005).
Philipson, L.H. & Roe, M.W. When BAD is good for beta cells. Cell Metab 7, 280–1 (2008).

Save book to Kindle

To save this book to your Kindle, first ensure [email protected] is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×