全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Cells  2013 

Linking Metabolic Abnormalities to Apoptotic Pathways in Beta Cells in Type 2 Diabetes

DOI: 10.3390/cells2020266

Keywords: Type 2 diabetes, pancreatic beta cell, apoptosis, endoplasmic reticulum stress, oxidative stress, NLRP3 inflammasome, Bcl-2 pathway

Full-Text   Cite this paper   Add to My Lib

Abstract:

Pancreatic beta-cell apoptosis is an important feature of islets in type 2 diabetes. Apoptosis can occur through two major pathways, the extrinsic or death receptor mediated pathway, and the intrinsic or Bcl-2-regulated pathway. Hyperglycaemia, hyperlipidaemia and islet amyloid poly-peptide (IAPP) represent important possible causes of increased beta-cell apoptosis. Hyperglycaemia induces islet-cell apoptosis by the intrinsic pathway involving molecules of the Bcl-2 family. High concentrations of palmitate also activate intrinsic apoptosis in islets cells. IAPP oligomers can induce apoptosis by both intrinsic and extrinsic pathways. IL-1b produced through NLRP3 inflammasome activation can also induce islet cell death. Activation of the NLRP3 inflammasome may not be important for glucose or palmitate induced apoptosis in islets but may be important for IAPP mediated cell death. Endoplasmic reticulum (ER) and oxidative stress have been observed in beta cells in type 2 diabetes, and these could be the link between upstream metabolic abnormalities and downstream apoptotic machinery.

References

[1]  Butler, A.E.; Janson, J.; Bonner-Weir, S.; Ritzel, R.; Rizza, R.A.; Butler, P.C. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 2003, 52, 102–110, doi:10.2337/diabetes.52.1.102.
[2]  Clark, A.; Wells, C.A.; Buley, I.D.; Cruickshank, J.K.; Vanhegan, R.I.; Matthews, D.R.; Cooper, G.J.; Holman, R.R.; Turner, R.C. Islet amyloid, Increased A-cells, Reduced B-cells and exocrine fibrosis: Quantitative changes in the pancreas in type 2 diabetes. Diabetes Res. 1988, 9, 151–159.
[3]  Sakuraba, H.; Mizukami, H.; Yagihashi, N.; Wada, R.; Hanyu, C.; Yagihashi, S. Reduced beta-cell mass and expression of oxidative stress-related DNA damage in the islet of Japanese Type II diabetic patients. Diabetologia 2002, 45, 85–96, doi:10.1007/s125-002-8248-z.
[4]  Song, B.; Scheuner, D.; Ron, D.; Pennathur, S.; Kaufman, R.J. Chop deletion reduces oxidative stress, Improves beta cell function, and promotes cell survival in multiple mouse models of diabetes. J. Clin. Invest. 2008, 118, 3378–3389.
[5]  Oyadomari, S.; Koizumi, A.; Takeda, K.; Gotoh, T.; Akira, S.; Araki, E.; Mori, M. Targeted disruption of the Chop gene delays endoplasmic reticulum stress-mediated diabetes. J. Clin. Invest. 2002, 109, 525–532.
[6]  Donath, M.Y.; Gross, D.J.; Cerasi, E.; Kaiser, N. Hyperglycemia-induced beta-cell apoptosis in pancreatic islets of Psammomys obesus during development of diabetes. Diabetes 1999, 48, 738–744, doi:10.2337/diabetes.48.4.738.
[7]  Pick, A.; Clark, J.; Kubstrup, C.; Levisetti, M.; Pugh, W.; Bonner-Weir, S.; Polonsky, K.S. 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, 358–364.
[8]  Zini, E.; Osto, M.; Franchini, M.; Guscetti, F.; Donath, M.Y.; Perren, A.; Heller, R.S.; Linscheid, P.; Bouwman, M.; Ackermann, M.; et al. Hyperglycaemia but not hyperlipidaemia causes beta cell dysfunction and beta cell loss in the domestic cat. Diabetologia 2009, 52, 336–346, doi:10.1007/s00125-008-1201-y.
[9]  Hotchkiss, R.S.; Strasser, A.; McDunn, J.E.; Swanson, P.E. Cell death. New Engl. J. Med. 2009, 361, 1570–1583, doi:10.1056/NEJMra0901217.
[10]  Strasser, A. The role of BH3-only proteins in the immune system. Nat. Rev. Immunol. 2005, 5, 189–200, doi:10.1038/nri1568.
[11]  Thomas, H.E.; McKenzie, M.D.; Angstetra, E.; Campbell, P.D.; Kay, T.W. Beta cell apoptosis in diabetes. Apoptosis 2009, 14, 1389–1404, doi:10.1007/s10495-009-0339-5.
[12]  Bergmann, L.; Kroncke, K.D.; Suschek, C.; Kolb, H.; Kolb-Bachofern, V. Cytotoxic action of IL-1 beta against pancreatic islets is mediated via nitric oxide formation and is inhibited by NG-monomethyl-L-arginine. FEBS Lett. 1992, 299, 103–106.
[13]  Maedler, K.; Sergeev, P.; Ris, F.; Oberholzer, J.; Joller-Jemelka, H.I.; Spinas, G.A.; Kaiser, N.; Halban, P.A.; Donath, M.Y. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J. Clin. Invest. 2002, 110, 851–860.
[14]  Masters, S.L.; Dunne, A.; Subramanian, S.L.; Hull, R.L.; Tannahill, G.M.; Sharp, F.A.; Becker, C.; Franchi, L.; Yoshihara, E.; Chen, Z.; et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1beta in type 2 diabetes. Nat. Immunol. 2010, 11, 897–904, doi:10.1038/ni.1935.
[15]  Ichinohe, T.; Pang, I.K.; Kumamoto, Y.; Peaper, D.R.; Ho, J.H.; Murray, T.S.; Iwasaki, A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc. Natl. Acad. Sci. USA 2011, 108, 5354–5359.
[16]  Joosten, L.A.; Netea, M.G.; Mylona, E.; Koenders, M.I.; Malireddi, R.K.; Oosting, M.; Stienstra, R.; van de Veerdonk, F.L.; Stalenhoef, A.F.; Giamarellos-Bourboulis, E.J.; et al. Engagement of fatty acids with Toll-like receptor 2 drives interleukin-1beta production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 2010, 62, 3237–3248, doi:10.1002/art.27667.
[17]  Lerner, A.G.; Upton, J.P.; Praveen, P.V.; Ghosh, R.; Nakagawa, Y.; Igbaria, A.; Shen, S.; Nguyen, V.; Backes, B.J.; Heiman, M.; et al. IRE1alpha induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell. Metab. 2012, 16, 250–264, doi:10.1016/j.cmet.2012.07.007.
[18]  Zhou, R.; Tardivel, A.; Thorens, B.; Choi, I.; Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 2010, 11, 136–140, doi:10.1038/ni.1831.
[19]  Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225.
[20]  Oslowski, C.M.; Hara, T.; O'Sullivan-Murphy, B.; Kanekura, K.; Lu, S.; Hara, M.; Ishigaki, S.; Zhu, L.J.; Hayashi, E.; Hui, S.T.; et al. Thioredoxin-interacting protein mediates ER stress-induced beta cell death through initiation of the inflammasome. Cell. Metab. 2012, 16, 265–273, doi:10.1016/j.cmet.2012.07.005.
[21]  Mahr, S.; Neumayer, N.; Gerhard, M.; Classen, M.; Prinz, C. IL-1beta-induced apoptosis in rat gastric enterochromaffin-like cells is mediated by iNOS, NF-kappaB, and Bax protein. Gastroenterology 2000, 118, 515–524.
[22]  Storling, J.; Binzer, J.; Andersson, A.K.; Zullig, R.A.; Tonnesen, M.; Lehmann, R.; Spinas, G.A.; Sandler, S.; Billestrup, N.; Mandrup-Poulsen, T. Nitric oxide contributes to cytokine-induced apoptosis in pancreatic beta cells via potentiation of JNK activity and inhibition of Akt. Diabetologia 2005, 48, 2039–2050, doi:10.1007/s00125-005-1912-2.
[23]  Thomas, H.E.; Darwiche, R.; Corbett, J.A.; Kay, T.W. Interleukin-1 plus gamma-interferon-induced pancreatic beta-cell dysfunction is mediated by beta-cell nitric oxide production. Diabetes 2002, 51, 311–316.
[24]  Kepp, O.; Galluzzi, L.; Zitvogel, L.; Kroemer, G. Pyroptosis — a cell death modality of its kind? Eur. J. Immunol. 2010, 40, 627–630, doi:10.1002/eji.200940160.
[25]  McKenzie, M.D.; Jamieson, E.; Jansen, E.S.; Scott, C.L.; Huang, D.C.; Bouillet, P.; Allison, J.; Kay, T.W.; Strasser, A.; Thomas, H.E. Glucose induces pancreatic islet cell apoptosis that requires the BH3-only proteins bim and puma and multi-BH domain protein Bax. Diabetes 2010, 59, 644–652, doi:10.2337/db09-1151.
[26]  Mellado-Gil, J.M.; Aguilar-Diosdado, M. High glucose potentiates cytokine- and streptozotocin-induced apoptosis of rat islet cells: Effect on apoptosis-related genes. J. Endocrinol. 2004, 183, 155–162, doi:10.1677/joe.1.05542.
[27]  Piro, S.; Anello, M.; Di Pietro, C.; Lizzio, M.N.; Patane, G.; Rabuazzo, A.M.; Vigneri, R.; Purrello, M.; Purrello, F. Chronic exposure to free fatty acids or high glucose induces apoptosis in rat pancreatic islets: possible role of oxidative stress. Metabolism 2002, 51, 1340–1347, doi:10.1053/meta.2002.35200.
[28]  Federici, M.; Hribal, M.; Perego, L.; Ranalli, M.; Caradonna, Z.; Perego, C.; Usellini, L.; Nano, R.; Bonini, P.; Bertuzzi, F.; et al. High glucose causes apoptosis in cultured human pancreatic islets of Langerhans: a potential role for regulation of specific Bcl family genes toward an apoptotic cell death program. Diabetes 2001, 50, 1290–1301, doi:10.2337/diabetes.50.6.1290.
[29]  Maedler, K.; Spinas, G.A.; Lehmann, R.; Sergeev, P.; Weber, M.; Fontana, A.; Kaiser, N.; Donath, M.Y. Glucose induces beta-cell apoptosis via upregulation of the Fas receptor in human islets. Diabetes 2001, 50, 1683–1690, doi:10.2337/diabetes.50.8.1683.
[30]  McKenzie, M.D.; Carrington, E.M.; Kaufmann, T.; Strasser, A.; Huang, D.C.; Kay, T.W.; Allison, J.; Thomas, H.E. Proapoptotic BH3-only protein Bid is essential for death receptor-induced apoptosis of pancreatic beta-cells. Diabetes 2008, 57, 1284–1292.
[31]  Westerbacka, J.; Lammi, K.; Hakkinen, A.M.; Rissanen, A.; Salminen, I.; Aro, A.; Yki-Jarvinen, H. Dietary fat content modifies liver fat in overweight nondiabetic subjects. J. Clin. Endocrinol. Metab. 2005, 90, 2804–2809, doi:10.1210/jc.2004-1983.
[32]  Cunha, D.A.; Hekerman, P.; Ladriere, L.; Bazarra-Castro, A.; Ortis, F.; Wakeham, M.C.; Moore, F.; Rasschaert, J.; Cardozo, A.K.; Bellomo, E.; et al. Initiation and execution of lipotoxic ER stress in pancreatic beta-cells. J. Cell Sci. 2008, 121, 2308–2318, doi:10.1242/jcs.026062.
[33]  Johnson, J.D.; Han, Z.; Otani, K.; Ye, H.; Zhang, Y.; Wu, H.; Horikawa, Y.; Misler, S.; Bell, G.I.; Polonsky, K.S. RyR2 and calpain-10 delineate a novel apoptosis pathway in pancreatic islets. J. Biol. Chem. 2004, 279, 24794–24802, doi:10.1074/jbc.M401216200.
[34]  Laybutt, D.R.; Preston, A.M.; Akerfeldt, M.C.; Kench, J.G.; Busch, A.K.; Biankin, A.V.; Biden, T.J. Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 2007, 50, 752–763, doi:10.1007/s00125-006-0590-z.
[35]  Cunha, D.A.; Igoillo-Esteve, M.; Gurzov, E.N.; Germano, C.M.; Naamane, N.; Marhfour, I.; Fukaya, M.; Vanderwinden, J.M.; Gysemans, C.; Mathieu, C.; et al. Death protein 5 and p53-upregulated modulator of apoptosis mediate the endoplasmic reticulum stress-mitochondrial dialog triggering lipotoxic rodent and human beta-cell apoptosis. Diabetes 2012, 61, 2763–2775, doi:10.2337/db12-0123.
[36]  Wen, H.; Gris, D.; Lei, Y.; Jha, S.; Zhang, L.; Huang, M.T.; Brickey, W.J.; Ting, J.P. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 2011, 12, 408–415, doi:10.1038/ni.2022.
[37]  Montane, J.; Klimek-Abercrombie, A.; Potter, K.J.; Westwell-Roper, C.; Bruce Verchere, C. Metabolic stress, IAPP and islet amyloid. Diabetes Obes. Metab. 2012, 14 (Suppl. 3), 68–77, doi:10.1111/j.1463-1326.2012.01657.x.
[38]  Westermark, P.; Andersson, A.; Westermark, G.T. Islet amyloid polypeptide, Islet amyloid, and diabetes mellitus. Physiol. Rev. 2011, 91, 795–826, doi:10.1152/physrev.00042.2009.
[39]  Zraika, S.; Hull, R.L.; Verchere, C.B.; Clark, A.; Potter, K.J.; Fraser, P.E.; Raleigh, D.P.; Kahn, S.E. Toxic oligomers and islet beta cell death: Guilty by association or convicted by circumstantial evidence? Diabetologia 2010, 53, 1046–1056, doi:10.1007/s00125-010-1671-6.
[40]  Jurgens, C.A.; Toukatly, M.N.; Fligner, C.L.; Udayasankar, J.; Subramanian, S.L.; Zraika, S.; Aston-Mourney, K.; Carr, D.B.; Westermark, P.; Westermark, G.T.; et al. beta-cell loss and beta-cell apoptosis in human type 2 diabetes are related to islet amyloid deposition. Am. J. Pathol. 2011, 178, 2632–2640, doi:10.1016/j.ajpath.2011.02.036.
[41]  Ritzel, R.A.; Meier, J.J.; Lin, C.Y.; Veldhuis, J.D.; Butler, P.C. Human islet amyloid polypeptide oligomers disrupt cell coupling, Induce apoptosis, and impair insulin secretion in isolated human islets. Diabetes 2007, 56, 65–71, doi:10.2337/db06-0734.
[42]  Janson, J.; Ashley, R.H.; Harrison, D.; McIntyre, S.; Butler, P.C. The mechanism of islet amyloid polypeptide toxicity is membrane disruption by intermediate-sized toxic amyloid particles. Diabetes 1999, 48, 491–498, doi:10.2337/diabetes.48.3.491.
[43]  Subramanian, S.L.; Hull, R.L.; Zraika, S.; Aston-Mourney, K.; Udayasankar, J.; Kahn, S.E. cJUN N-terminal kinase (JNK) activation mediates islet amyloid-induced beta cell apoptosis in cultured human islet amyloid polypeptide transgenic mouse islets. Diabetologia 2012, 55, 166–174, doi:10.1007/s00125-011-2338-7.
[44]  Park, Y.J.; Lee, S.; Kieffer, T.J.; Warnock, G.L.; Safikhan, N.; Speck, M.; Hao, Z.; Woo, M.; Marzban, L. Deletion of Fas protects islet beta cells from cytotoxic effects of human islet amyloid polypeptide. Diabetologia 2012, 55, 1035–1047, doi:10.1007/s00125-012-2451-2.
[45]  Westwell-Roper, C.; Dai, D.L.; Soukhatcheva, G.; Potter, K.J.; van Rooijen, N.; Ehses, J.A.; Verchere, C.B. IL-1 blockade attenuates islet amyloid polypeptide-induced proinflammatory cytokine release and pancreatic islet graft dysfunction. J. Immunol. 2011, 187, 2755–2765, doi:10.4049/jimmunol.1002854.
[46]  Eizirik, D.L.; Cardozo, A.K.; Cnop, M. The role for endoplasmic reticulum stress in diabetes mellitus. Endocr. Rev. 2008, 29, 42–61, doi:10.1210/er.2007-0015.
[47]  Fonseca, S.G.; Burcin, M.; Gromada, J.; Urano, F. Endoplasmic reticulum stress in beta-cells and development of diabetes. Curr. Opin. Pharmacol. 2009, 9, 763–770.
[48]  Fonseca, S.G.; Gromada, J.; Urano, F. Endoplasmic reticulum stress and pancreatic beta-cell death. Trends Endocrinol. Metab. 2011, 22, 266–274.
[49]  Lipson, K.L.; Ghosh, R.; Urano, F. The role of IRE1alpha in the degradation of insulin mRNA in pancreatic beta-cells. PLoS One 2008, 3, e1648, doi:10.1371/journal.pone.0001648.
[50]  Hou, Z.Q.; Li, H.L.; Gao, L.; Pan, L.; Zhao, J.J.; Li, G.W. Involvement of chronic stresses in rat islet and INS-1 cell glucotoxicity induced by intermittent high glucose. Mol. Cell. Endocrinol. 2008, 291, 71–78.
[51]  Maedler, K.; Schulthess, F.T.; Bielman, C.; Berney, T.; Bonny, C.; Prentki, M.; Donath, M.Y.; Roduit, R. Glucose and leptin induce apoptosis in human beta-cells and impair glucose-stimulated insulin secretion through activation of c-Jun N-terminal kinases. FASEB J. 2008, 22, 1905–1913, doi:10.1096/fj.07-101824.
[52]  Hull, R.L.; Zraika, S.; Udayasankar, J.; Aston-Mourney, K.; Subramanian, S.L.; Kahn, S.E. Amyloid formation in human IAPP transgenic mouse islets and pancreas, and human pancreas, is not associated with endoplasmic reticulum stress. Diabetologia 2009, 52, 1102–1111, doi:10.1007/s00125-009-1329-4.
[53]  Matveyenko, A.V.; Gurlo, T.; Daval, M.; Butler, A.E.; Butler, P.C. Successful versus failed adaptation to high-fat diet-induced insulin resistance: the role of IAPP-induced beta-cell endoplasmic reticulum stress. Diabetes 2009, 58, 906–916, doi:10.2337/db08-1464.
[54]  Huang, C.J.; Lin, C.Y.; Haataja, L.; Gurlo, T.; Butler, A.E.; Rizza, R.A.; Butler, P.C. 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, 2016–2027, doi:10.2337/db07-0197.
[55]  Cnop, M.; Foufelle, F.; Velloso, L.A. Endoplasmic reticulum stress, obesity and diabetes. Trends Mol. Med. 2012, 18, 59–68, doi:10.1016/j.molmed.2011.07.010.
[56]  Puthalakath, H.; O'Reilly, L.A.; Gunn, P.; Lee, L.; Kelly, P.N.; Huntington, N.D.; Hughes, P.D.; Michalak, E.M.; McKimm-Breschkin, J.; Motoyama, N.; et al. ER stress triggers apoptosis by activating BH3-only protein Bim. Cell 2007, 129, 1337–1349, doi:10.1016/j.cell.2007.04.027.
[57]  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. 2006, 281, 7260–7270, doi:10.1074/jbc.M509868200.
[58]  Kieran, D.; Woods, I.; Villunger, A.; Strasser, A.; Prehn, J.H. Deletion of the BH3-only protein puma protects motoneurons from ER stress-induced apoptosis and delays motoneuron loss in ALS mice. Proc. Natl. Acad. Sci. USA 2007, 104, 20606–20611.
[59]  Cazanave, S.C.; Elmi, N.A.; Akazawa, Y.; Bronk, S.F.; Mott, J.L.; Gores, G.J. CHOP and AP-1 cooperatively mediate PUMA expression during lipoapoptosis. Am. J. Physiol.-Gastr. L. 2010, 299, G236–G243.
[60]  Giam, M.; Huang, D.C.; Bouillet, P. BH3-only proteins and their roles in programmed cell death. Oncogene 2008, 27 (Suppl. 1), S128–S136, doi:10.1038/onc.2009.50.
[61]  Steckley, D.; Karajgikar, M.; Dale, L.B.; Fuerth, B.; Swan, P.; Drummond-Main, C.; Poulter, M.O.; Ferguson, S.S.; Strasser, A.; Cregan, S.P. Puma is a dominant regulator of oxidative stress induced Bax activation and neuronal apoptosis. J. Neurosci. 2007, 27, 12989–12999, doi:10.1523/JNEUROSCI.3400-07.2007.
[62]  Ghosh, A.P.; Klocke, B.J.; Ballestas, M.E.; Roth, K.A. CHOP potentially co-operates with FOXO3a in neuronal cells to regulate PUMA and BIM expression in response to ER stress. PLoS One 2012, 7, e39586.
[63]  Menu, P.; Mayor, A.; Zhou, R.; Tardivel, A.; Ichijo, H.; Mori, K.; Tschopp, J. ER stress activates the NLRP3 inflammasome via an UPR-independent pathway. Cell. Death Dis. 2012, 3, e261.
[64]  Chen, J.; Fontes, G.; Saxena, G.; Poitout, V.; Shalev, A. Lack of TXNIP protects against mitochondria-mediated apoptosis but not against fatty acid-induced ER stress-mediated beta-cell death. Diabetes 2010, 59, 440–447, doi:10.2337/db09-0949.
[65]  Mates, J.M. Effects of antioxidant enzymes in the molecular control of reactive oxygen species toxicology. Toxicology 2000, 153, 83–104, doi:10.1016/S0300-483X(00)00306-1.
[66]  Jonas, J.C.; Bensellam, M.; Duprez, J.; Elouil, H.; Guiot, Y.; Pascal, S.M. Glucose regulation of islet stress responses and beta-cell failure in type 2 diabetes. Diabetes Obes. Metab. 2009, 11 (Suppl. 4), 65–81, doi:10.1111/j.1463-1326.2009.01112.x.
[67]  Lenzen, S.; Drinkgern, J.; Tiedge, M. Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues. Free Radical Biol. Med. 1996, 20, 463–466, doi:10.1016/0891-5849(96)02051-5.
[68]  Moore, P.C.; Ugas, M.A.; Hagman, D.K.; Parazzoli, S.D.; Poitout, V. Evidence against the involvement of oxidative stress in fatty acid inhibition of insulin secretion. Diabetes 2004, 53, 2610–2616, doi:10.2337/diabetes.53.10.2610.
[69]  Tanaka, Y.; Tran, P.O.; Harmon, J.; Robertson, R.P. A role for glutathione peroxidase in protecting pancreatic beta cells against oxidative stress in a model of glucose toxicity. Proc. Natl. Acad. Sci. USA 2002, 99, 12363–12368.
[70]  Kaneto, H.; Kajimoto, Y.; Miyagawa, J.; Matsuoka, T.; Fujitani, Y.; Umayahara, Y.; Hanafusa, T.; Matsuzawa, Y.; Yamasaki, Y.; Hori, M. Beneficial effects of antioxidants in diabetes: possible protection of pancreatic beta-cells against glucose toxicity. Diabetes 1999, 48, 2398–2406, doi:10.2337/diabetes.48.12.2398.
[71]  Oprescu, A.I.; Bikopoulos, G.; Naassan, A.; Allister, E.M.; Tang, C.; Park, E.; Uchino, H.; Lewis, G.F.; Fantus, I.G.; Rozakis-Adcock, M.; et al. Free fatty acid-induced reduction in glucose-stimulated insulin secretion: Evidence for a role of oxidative stress in vitro and in vivo. Diabetes 2007, 56, 2927–2937, doi:10.2337/db07-0075.
[72]  Carlsson, C.; Borg, L.A.; Welsh, N. Sodium palmitate induces partial mitochondrial uncoupling and reactive oxygen species in rat pancreatic islets in vitro. Endocrinology 1999, 140, 3422–3428, doi:10.1210/en.140.8.3422.
[73]  Zraika, S.; Hull, R.L.; Udayasankar, J.; Aston-Mourney, K.; Subramanian, S.L.; Kisilevsky, R.; Szarek, W.A.; Kahn, S.E. Oxidative stress is induced by islet amyloid formation and time-dependently mediates amyloid-induced beta cell apoptosis. Diabetologia 2009, 52, 626–635, doi:10.1007/s00125-008-1255-x.
[74]  Jiang, D.; Jha, N.; Boonplueang, R.; Andersen, J.K. Caspase 3 inhibition attenuates hydrogen peroxide-induced DNA fragmentation but not cell death in neuronal PC12 cells. J. Neuro. Chem. 2001, 76, 1745–1755.
[75]  Takahashi, A.; Masuda, A.; Sun, M.; Centonze, V.E.; Herman, B. Oxidative stress-induced apoptosis is associated with alterations in mitochondrial caspase activity and Bcl-2-dependent alterations in mitochondrial pH (pHm). Brain Res. Bull. 2004, 62, 497–504, doi:10.1016/j.brainresbull.2003.07.009.
[76]  Cai, Y.; Martens, G.A.; Hinke, S.A.; Heimberg, H.; Pipeleers, D.; Van de Casteele, M. Increased oxygen radical formation and mitochondrial dysfunction mediate beta cell apoptosis under conditions of AMP-activated protein kinase stimulation. Free Radical Biol. Med. 2007, 42, 64–78, doi:10.1016/j.freeradbiomed.2006.09.018.
[77]  Wang, M.; Crager, M.; Pugazhenthi, S. Modulation of apoptosis pathways by oxidative stress and autophagy in beta cells. Exp. Diabetes Res. 2012, 2012, 647914.
[78]  Petrilli, V.; Papin, S.; Dostert, C.; Mayor, A.; Martinon, F.; Tschopp, J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell. Death Differ. 2007, 14, 1583–1589, doi:10.1038/sj.cdd.4402195.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413