Redox Homeostasis in Pancreatic β-Cells: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Blanka Holendova.

Redox status is a key determinant in the fate of every cell, β-cell in particular.  β-cells are not primarily detoxifying like e.g. hepatocytes or kidney cells and thus do not possess extensive antioxidant defense machinery. However, they show a wide range of redox regulating proteins, such as peroxiredoxins, thioredoxins or thioredoxin reductases, etc., being functionally compartmentalized. These proteins keep fragile redox homeostasis and serve as messengers and amplifiers of redox signaling which is inevitable for proper β-cell function and maintanance.

Redox status is a key determinant in the fate of every cell, β-cell in particular.  β-cells are not primarily detoxifying like e.g. hepatocytes or kidney cells and thus do not possess extensive antioxidant defense machinery. However, they show a wide range of redox regulating proteins, such as peroxiredoxins, thioredoxins or thioredoxin reductases, etc., being functionally compartmentalized. These proteins keep fragile redox homeostasis and serve as messengers and amplifiers of redox signaling which is inevitable for proper β-cell function and maintanance.

  • redox signaling
  • oxidative stress
  • redox homeostasis
  • pancreatic β-cells
Please wait, diff process is still running!

References

  1. Leloup, C.; Tourrel-Cuzin, C.; Magnan, C.; Karaca, M.; Castel, J.; Carneiro, L.; Colombani, A.L.; Ktorza, A.; Casteilla, L.; Penicaud, L. Mitochondrial reactive oxygen species are obligatory signals for glucose-induced insulin secretion. Diabetes 2009, 58, 673–681.
  2. Plecita-Hlavata, L.; Jaburek, M.; Holendova, B.; Tauber, J.; Pavluch, V.; Berkova, Z.; Cahova, M.; Schroeder, K.; Brandes, R.P.; Siemen, D.; et al. Glucose-Stimulated Insulin Secretion Fundamentally Requires H2O2 Signaling by NADPH Oxidase 4. Diabetes 2020.
  3. Wang, J.; Wang, H. Oxidative Stress in Pancreatic Beta Cell Regeneration. Oxid. Med. Cell. Longev. 2017, 2017, 1930261.
  4. 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. 1981, 199, 393–398.
  5. Lenzen, S.; Drinkgern, J.; Tiedge, M. Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues. Free Radic. Biol. Med. 1996, 20, 463–466.
  6. Tiedge, M.; Lortz, S.; Drinkgern, J.; Lenzen, S. Relation between antioxidant enzyme gene expression and antioxidative defense status of insulin-producing cells. Diabetes 1997, 46, 1733–1742.
  7. Miki, A.; Ricordi, C.; Sakuma, Y.; Yamamoto, T.; Misawa, R.; Mita, A.; Molano, R.D.; Vaziri, N.D.; Pileggi, A.; Ichii, H. Divergent antioxidant capacity of human islet cell subsets: A potential cause of beta-cell vulnerability in diabetes and islet transplantation. PLoS ONE 2018, 13, e0196570.
  8. Kalinina, E.V.; Chernov, N.N.; Saprin, A.N. Involvement of thio-, peroxi-, and glutaredoxins in cellular redox-dependent processes. Biochemistry 2008, 73, 1493–1510.
  9. Stancill, J.S.; Broniowska, K.A.; Oleson, B.J.; Naatz, A.; Corbett, J.A. Pancreatic beta-cells detoxify H2O2 through the peroxiredoxin/thioredoxin antioxidant system. J. Biol. Chem. 2019, 294, 4843–4853.
  10. Munro, D.; Treberg, J.R. A radical shift in perspective: Mitochondria as regulators of reactive oxygen species. J. Exp. Biol. 2017, 220, 1170–1180.
  11. Roma, L.P.; Jonas, J.C. Nutrient Metabolism, Subcellular Redox State, and Oxidative Stress in Pancreatic Islets and beta-Cells. J. Mol. Biol. 2019.
  12. Yoboue, E.D.; Sitia, R.; Simmen, T. Redox crosstalk at endoplasmic reticulum (ER) membrane contact sites (MCS) uses toxic waste to deliver messages. Cell Death Dis. 2018, 9, 331.
  13. Gurgul, E.; Lortz, S.; Tiedge, M.; Jorns, A.; Lenzen, S. Mitochondrial catalase overexpression protects insulin-producing cells against toxicity of reactive oxygen species and proinflammatory cytokines. Diabetes 2004, 53, 2271–2280.
  14. Plecita-Hlavata, L.; Engstova, H.; Jezek, J.; Holendova, B.; Tauber, J.; Petraskova, L.; Kren, V.; Jezek, P. Potential of Mitochondria-Targeted Antioxidants to Prevent Oxidative Stress in Pancreatic beta-cells. Oxid. Med. Cell. Longev. 2019, 2019, 1826303.
  15. Plecita-Hlavata, L.; Engstova, H.; Holendova, B.; Tauber, J.; Spacek, T.; Petraskova, L.; Kren, V.; Spackova, J.; Gotvaldova, K.; Jezek, J.; et al. Mitochondrial Superoxide Production Decreases on Glucose-Stimulated Insulin Secretion in Pancreatic beta Cells Due to Decreasing Mitochondrial Matrix NADH/NAD(+) Ratio. Antioxid. Redox Signal. 2020, 33, 789–815.
  16. Jezek, J.; Dlaskova, A.; Zelenka, J.; Jaburek, M.; Jezek, P. H(2)O(2)-Activated Mitochondrial Phospholipase iPLA(2)gamma Prevents Lipotoxic Oxidative Stress in Synergy with UCP2, Amplifies Signaling via G-Protein-Coupled Receptor GPR40, and Regulates Insulin Secretion in Pancreatic beta-Cells. Antioxid. Redox Signal. 2015, 23, 958–972.
  17. Jezek, P.; Holendova, B.; Garlid, K.D.; Jaburek, M. Mitochondrial Uncoupling Proteins: Subtle Regulators of Cellular Redox Signaling. Antioxid. Redox Signal. 2018, 29, 667–714.
  18. Appenzeller-Herzog, C.; Riemer, J.; Zito, E.; Chin, K.T.; Ron, D.; Spiess, M.; Ellgaard, L. Disulphide production by Ero1alpha-PDI relay is rapid and effectively regulated. EMBO J. 2010, 29, 3318–3329.
  19. Mehmeti, I.; Lortz, S.; Elsner, M.; Lenzen, S. Peroxiredoxin 4 improves insulin biosynthesis and glucose-induced insulin secretion in insulin-secreting INS-1E cells. J. Biol. Chem. 2014, 289, 26904–26913.
  20. Lenzen, S. Chemistry and biology of reactive species with special reference to the antioxidative defence status in pancreatic beta-cells. Biochim Biophys Acta Gen. Subj. 2017, 1861, 1929–1942.
  21. Tavender, T.J.; Sheppard, A.M.; Bulleid, N.J. Peroxiredoxin IV is an endoplasmic reticulum-localized enzyme forming oligomeric complexes in human cells. Biochem. J. 2008, 411, 191–199.
  22. Nguyen, V.D.; Saaranen, M.J.; Karala, A.R.; Lappi, A.K.; Wang, L.; Raykhel, I.B.; Alanen, H.I.; Salo, K.E.; Wang, C.C.; Ruddock, L.W. Two endoplasmic reticulum PDI peroxidases increase the efficiency of the use of peroxide during disulfide bond formation. J. Mol. Biol. 2011, 406, 503–515.
  23. Hassler, J.R.; Scheuner, D.L.; Wang, S.; Han, J.; Kodali, V.K.; Li, P.; Nguyen, J.; George, J.S.; Davis, C.; Wu, S.P.; et al. The IRE1alpha/XBP1s Pathway Is Essential for the Glucose Response and Protection of beta Cells. PLoS Biol. 2015, 13, e1002277.
  24. Pearse, B.R.; Hebert, D.N. Lectin chaperones help direct the maturation of glycoproteins in the endoplasmic reticulum. Biochim. Biophys. Acta 2010, 1803, 684–693.
  25. Plemper, R.K.; Bohmler, S.; Bordallo, J.; Sommer, T.; Wolf, D.H. Mutant analysis links the translocon and BiP to retrograde protein transport for ER degradation. Nature 1997, 388, 891–895.
  26. Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086.
  27. Bertolotti, A.; Zhang, Y.; Hendershot, L.M.; Harding, H.P.; Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat. Cell Biol. 2000, 2, 326–332.
  28. Brozzi, F.; Eizirik, D.L. ER stress and the decline and fall of pancreatic beta cells in type 1 diabetes. Ups J. Med. Sci. 2016, 121, 133–139.
  29. Baboota, R.K.; Shinde, A.B.; Lemaire, K.; Fransen, M.; Vinckier, S.; Van Veldhoven, P.P.; Schuit, F.; Baes, M. Functional peroxisomes are required for beta-cell integrity in mice. Mol. Metab. 2019, 22, 71–83.
  30. Oliveira, H.R.; Verlengia, R.; Carvalho, C.R.; Britto, L.R.; Curi, R.; Carpinelli, A.R. Pancreatic beta-cells express phagocyte-like NAD(P)H oxidase. Diabetes 2003, 52, 1457–1463.
  31. Uchizono, Y.; Takeya, R.; Iwase, M.; Sasaki, N.; Oku, M.; Imoto, H.; Iida, M.; Sumimoto, H. Expression of isoforms of NADPH oxidase components in rat pancreatic islets. Life Sci. 2006, 80, 133–139.
  32. Zhang, Z.; Li, J.; Yang, L.; Chen, R.; Yang, R.; Zhang, H.; Cai, D.; Chen, H. The cytotoxic role of intermittent high glucose on apoptosis and cell viability in pancreatic beta cells. J. Diabetes Res. 2014, 2014, 712781.
  33. Stancill, J.S.; Happ, J.T.; Broniowska, K.A.; Hogg, N.; Corbett, J.A. Peroxiredoxin 1 plays a primary role in protecting pancreatic β-cells from hydrogen peroxide and peroxynitrite. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2020, 318, R1004–R1013.
  34. Nishiyama, A.; Matsui, M.; Iwata, S.; Hirota, K.; Masutani, H.; Nakamura, H.; Takagi, Y.; Sono, H.; Gon, Y.; Yodoi, J. Identification of thioredoxin-binding protein-2/vitamin D(3) up-regulated protein 1 as a negative regulator of thioredoxin function and expression. J. Biol. Chem. 1999, 274, 21645–21650.
  35. Wondafrash, D.Z.; Nire’a, A.T.; Tafere, G.G.; Desta, D.M.; Berhe, D.A.; Zewdie, K.A. Thioredoxin-Interacting Protein as a Novel Potential Therapeutic Target in Diabetes Mellitus and Its Underlying Complications. Diabetes Metab. Syndr. Obes. 2020, 13, 43–51.
  36. Yoshihara, E.; Masaki, S.; Matsuo, Y.; Chen, Z.; Tian, H.; Yodoi, J. Thioredoxin/Txnip: Redoxisome, as a redox switch for the pathogenesis of diseases. Front. Immunol. 2014, 4, 514.
  37. Nishinaka, Y.; Masutani, H.; Oka, S.; Matsuo, Y.; Yamaguchi, Y.; Nishio, K.; Ishii, Y.; Yodoi, J. Importin alpha1 (Rch1) mediates nuclear translocation of thioredoxin-binding protein-2/vitamin D(3)-up-regulated protein 1. J. Biol. Chem. 2004, 279, 37559–37565.
  38. Xu, G.; Chen, J.; Jing, G.; Shalev, A. Thioredoxin-interacting protein regulates insulin transcription through microRNA-204. Nat. Med. 2013, 19, 1141–1146.
  39. Jing, G.; Westwell-Roper, C.; Chen, J.; Xu, G.; Verchere, C.B.; Shalev, A. Thioredoxin-interacting protein promotes islet amyloid polypeptide expression through miR-124a and FoxA2. J. Biol. Chem. 2014, 289, 11807–11815.
  40. Pi, J.; Zhang, Q.; Fu, J.; Woods, C.G.; Hou, Y.; Corkey, B.E.; Collins, S.; Andersen, M.E. ROS signaling, oxidative stress and Nrf2 in pancreatic beta-cell function. Toxicol. Appl. Pharm. 2010, 244, 77–83.
  41. Janjic, D.; Maechler, P.; Sekine, N.; Bartley, C.; Annen, A.S.; Wolheim, C.B. Free radical modulation of insulin release in INS-1 cells exposed to alloxan. Biochem. Pharm. 1999, 57, 639–648.
  42. Maechler, P.; Jornot, L.; Wollheim, C.B. Hydrogen peroxide alters mitochondrial activation and insulin secretion in pancreatic beta cells. J. Biol. Chem. 1999, 274, 27905–27913.
  43. Pi, J.; Bai, Y.; Zhang, Q.; Wong, V.; Floering, L.M.; Daniel, K.; Reece, J.M.; Deeney, J.T.; Andersen, M.E.; Corkey, B.E.; et al. Reactive oxygen species as a signal in glucose-stimulated insulin secretion. Diabetes 2007, 56, 1783–1791.
  44. Travasso, R.D.M.; Sampaio Dos Aidos, F.; Bayani, A.; Abranches, P.; Salvador, A. Localized redox relays as a privileged mode of cytoplasmic hydrogen peroxide signaling. Redox Biol. 2017, 12, 233–245.
  45. Hanschmann, E.M.; Godoy, J.R.; Berndt, C.; Hudemann, C.; Lillig, C.H. Thioredoxins, glutaredoxins, and peroxiredoxins--molecular mechanisms and health significance: From cofactors to antioxidants to redox signaling. Antioxid. Redox Signal. 2013, 19, 1539–1605.
  46. Kontou, M.; Will, R.D.; Adelfalk, C.; Wittig, R.; Poustka, A.; Hirsch-Kauffmann, M.; Schweiger, M. Thioredoxin, a regulator of gene expression. Oncogene 2004, 23, 2146–2152.
  47. Bian, M.; Fan, R.; Zhao, S.; Liu, W. Targeting the Thioredoxin System as a Strategy for Cancer Therapy. J. Med. Chem. 2019, 62, 7309–7321.
  48. Jastrząb, A.; Skrzydlewska, E. Thioredoxin-dependent system. Application of inhibitors. J. Enzym. Inhib. Med. Chem. 2021, 36, 362–371.
  49. Muri, J.; Kopf, M. Redox regulation of immunometabolism. Nat. Reviews. Immunol. 2020.
  50. Zhang, J.; Wang, X.; Vikash, V.; Ye, Q.; Wu, D.; Liu, Y.; Dong, W. ROS and ROS-Mediated Cellular Signaling. Oxid. Med. Cell. Longev. 2016, 2016, 4350965.
  51. Schultheis, J.; Beckmann, D.; Mulac, D.; Muller, L.; Esselen, M.; Dufer, M. Nrf2 Activation Protects Mouse Beta Cells from Glucolipotoxicity by Restoring Mitochondrial Function and Physiological Redox Balance. Oxid. Med. Cell. Longev. 2019, 2019, 7518510.
  52. He, J.; Zhang, X.; Lian, C.; Wu, J.; Fang, Y.; Ye, X. KEAP1/NRF2 axis regulates H2O2-induced apoptosis of pancreatic beta-cells. Gene 2019, 691, 8–17.
  53. Cardozo, A.K.; Heimberg, H.; Heremans, Y.; Leeman, R.; Kutlu, B.; Kruhoffer, M.; Orntoft, T.; Eizirik, D.L. A comprehensive analysis of cytokine-induced and nuclear factor-kappa B-dependent genes in primary rat pancreatic beta-cells. J. Biol. Chem. 2001, 276, 48879–48886.
  54. Meyerovich, K.; Fukaya, M.; Terra, L.F.; Ortis, F.; Eizirik, D.L.; Cardozo, A.K. The non-canonical NF-kappaB pathway is induced by cytokines in pancreatic beta cells and contributes to cell death and proinflammatory responses in vitro. Diabetologia 2016, 59, 512–521.
  55. Meyerovich, K.; Ortis, F.; Cardozo, A.K. The non-canonical NF-kappaB pathway and its contribution to beta-cell failure in diabetes. J. Mol. Endocrinol. 2018, 61, F1–F6.
  56. Lee, K.; Esselman, W.J. Inhibition of PTPs by H(2)O(2) regulates the activation of distinct MAPK pathways. Free Radic. Biol. Med. 2002, 33, 1121–1132.
  57. Brigelius-Flohe, R.; Flohe, L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid. Redox Signal. 2011, 15, 2335–2381.
  58. Schulze-Osthoff, K.; Beyaert, R.; Vandevoorde, V.; Haegeman, G.; Fiers, W. Depletion of the mitochondrial electron transport abrogates the cytotoxic and gene-inductive effects of TNF. EMBO J. 1993, 12, 3095–3104.
  59. Baeuerle, P.A.; Henkel, T. Function and activation of NF-kappa B in the immune system. Annu. Rev. Immunol. 1994, 12, 141–179.
  60. Cerf, M.E. Transcription factors regulating beta-cell function. Eur. J. Endocrinol. 2006, 155, 671–679.
  61. Jara, M.A.; Werneck-De-Castro, J.P.; Lubaczeuski, C.; Johnson, J.D.; Bernal-Mizrachi, E. Pancreatic and duodenal homeobox-1 (PDX1) contributes to beta-cell mass expansion and proliferation induced by Akt/PKB pathway. Islets 2020, 12, 32–40.
  62. 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.
  63. Tanaka, Y.; Gleason, C.E.; Tran, P.O.; Harmon, J.S.; Robertson, R.P. Prevention of glucose toxicity in HIT-T15 cells and Zucker diabetic fatty rats by antioxidants. Proc. Natl. Acad. Sci. USA 1999, 96, 10857–10862.
  64. Matsuoka, T.A.; Kaneto, H.; Stein, R.; Miyatsuka, T.; Kawamori, D.; Henderson, E.; Kojima, I.; Matsuhisa, M.; Hori, M.; Yamasaki, Y. MafA regulates expression of genes important to islet beta-cell function. Mol. Endocrinol. 2007, 21, 2764–2774.
  65. Harmon, J.S.; Stein, R.; Robertson, R.P. Oxidative stress-mediated, post-translational loss of MafA protein as a contributing mechanism to loss of insulin gene expression in glucotoxic beta cells. J. Biol. Chem. 2005, 280, 11107–11113.
  66. Kondo, T.; El Khattabi, I.; Nishimura, W.; Laybutt, D.R.; Geraldes, P.; Shah, S.; King, G.; Bonner-Weir, S.; Weir, G.; Sharma, A. p38 MAPK is a major regulator of MafA protein stability under oxidative stress. Mol. Endocrinol. 2009, 23, 1281–1290.
  67. El Khattabi, I.; Sharma, A. Preventing p38 MAPK-mediated MafA degradation ameliorates beta-cell dysfunction under oxidative stress. Mol. Endocrinol. 2013, 27, 1078–1090.
  68. Harmon, J.S.; Bogdani, M.; Parazzoli, S.D.; Mak, S.S.; Oseid, E.A.; Berghmans, M.; Leboeuf, R.C.; Robertson, R.P. beta-Cell-specific overexpression of glutathione peroxidase preserves intranuclear MafA and reverses diabetes in db/db mice. Endocrinology 2009, 150, 4855–4862.
  69. Barthel, A.; Schmoll, D.; Unterman, T.G. FoxO proteins in insulin action and metabolism. Trends Endocrinol. Metab. 2005, 16, 183–189.
  70. Dansen, T.B.; Smits, L.M.; van Triest, M.H.; de Keizer, P.L.; van Leenen, D.; Koerkamp, M.G.; Szypowska, A.; Meppelink, A.; Brenkman, A.B.; Yodoi, J.; et al. Redox-sensitive cysteines bridge p300/CBP-mediated acetylation and FoxO4 activity. Nat. Chem. Biol. 2009, 5, 664–672.
  71. De Keizer, P.L.; Burgering, B.M.; Dansen, T.B. Forkhead box o as a sensor, mediator, and regulator of redox signaling. Antioxid. Redox Signal. 2011, 14, 1093–1106.
  72. Burgering, B.M.; Coffer, P.J. Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction. Nature 1995, 376, 599–602.
  73. Huang, X.; Begley, M.; Morgenstern, K.A.; Gu, Y.; Rose, P.; Zhao, H.; Zhu, X. Crystal structure of an inactive Akt2 kinase domain. Structure 2003, 11, 21–30.
  74. Murata, H.; Ihara, Y.; Nakamura, H.; Yodoi, J.; Sumikawa, K.; Kondo, T. Glutaredoxin exerts an antiapoptotic effect by regulating the redox state of Akt. J. Biol. Chem. 2003, 278, 50226–50233.
  75. Kitamura, T.; Nakae, J.; Kitamura, Y.; Kido, Y.; Biggs, W.H., 3rd; Wright, C.V.; White, M.F.; Arden, K.C.; Accili, D. The forkhead transcription factor Foxo1 links insulin signaling to Pdx1 regulation of pancreatic beta cell growth. J. Clin. Investig. 2002, 110, 1839–1847.
  76. Kitamura, Y.I.; Kitamura, T.; Kruse, J.P.; Raum, J.C.; Stein, R.; Gu, W.; Accili, D. FoxO1 protects against pancreatic beta cell failure through NeuroD and MafA induction. Cell Metab. 2005, 2, 153–163.
  77. Nishimura, W.; Kondo, T.; Salameh, T.; El Khattabi, I.; Dodge, R.; Bonner-Weir, S.; Sharma, A. A switch from MafB to MafA expression accompanies differentiation to pancreatic beta-cells. Dev. Biol. 2006, 293, 526–539.
  78. Cao, X.; Kambe, F.; Ohmori, S.; Seo, H. Oxidoreductive modification of two cysteine residues in paired domain by Ref-1 regulates DNA-binding activity of Pax-8. Biochem. Biophys Res. Commun 2002, 297, 288–293.
  79. Walther, C.; Guenet, J.L.; Simon, D.; Deutsch, U.; Jostes, B.; Goulding, M.D.; Plachov, D.; Balling, R.; Gruss, P. Pax: A murine multigene family of paired box-containing genes. Genomics 1991, 11, 424–434.
  80. Swisa, A.; Avrahami, D.; Eden, N.; Zhang, J.; Feleke, E.; Dahan, T.; Cohen-Tayar, Y.; Stolovich-Rain, M.; Kaestner, K.H.; Glaser, B.; et al. PAX6 maintains beta cell identity by repressing genes of alternative islet cell types. J. Clin. Investig. 2017, 127, 230–243.
  81. Rieck, S.; Bankaitis, E.D.; Wright, C.V. Lineage determinants in early endocrine development. In Seminars in Cell & Developmental Biology; Academic Press: Cambridge, MA, USA, 2012; Volume 23, pp. 673–684.
  82. Bastidas-Ponce, A.; Roscioni, S.S.; Burtscher, I.; Bader, E.; Sterr, M.; Bakhti, M.; Lickert, H. Foxa2 and Pdx1 cooperatively regulate postnatal maturation of pancreatic beta-cells. Mol. Metab. 2017, 6, 524–534.
  83. Bensellam, M.; Jonas, J.C.; Laybutt, D.R. Mechanisms of beta-cell dedifferentiation in diabetes: Recent findings and future research directions. J. Endocrinol. 2018, 236, R109–R143.
  84. Balakrishnan, S.; Dhavamani, S.; Prahalathan, C. beta-Cell specific transcription factors in the context of diabetes mellitus and beta-cell regeneration. Mech. Dev. 2020, 163, 103634.
  85. Zhou, Q.; Brown, J.; Kanarek, A.; Rajagopal, J.; Melton, D.A. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 2008, 455, 627–632.
  86. Chetboun, M.; Abitbol, G.; Rozenberg, K.; Rozenfeld, H.; Deutsch, A.; Sampson, S.R.; Rosenzweig, T. Maintenance of redox state and pancreatic beta-cell function: Role of leptin and adiponectin. J. Cell. Biochem. 2012, 113, 1966–1976.
  87. Ahmed Alfar, E.; Kirova, D.; Konantz, J.; Birke, S.; Mansfeld, J.; Ninov, N. Distinct Levels of Reactive Oxygen Species Coordinate Metabolic Activity with Beta-cell Mass Plasticity. Sci. Rep. 2017, 7, 3994.
  88. Liang, J.; Wu, S.Y.; Zhang, D.; Wang, L.; Leung, K.K.; Leung, P.S. NADPH Oxidase-Dependent Reactive Oxygen Species Stimulate beta-Cell Regeneration Through Differentiation of Endocrine Progenitors in Murine Pancreas. Antioxid. Redox Signal. 2016, 24, 419–433.
  89. Hoarau, E.; Chandra, V.; Rustin, P.; Scharfmann, R.; Duvillie, B. Pro-oxidant/antioxidant balance controls pancreatic beta-cell differentiation through the ERK1/2 pathway. Cell Death Dis. 2014, 5, e1487.
  90. Costes, S.; Broca, C.; Bertrand, G.; Lajoix, A.D.; Bataille, D.; Bockaert, J.; Dalle, S. ERK1/2 control phosphorylation and protein level of cAMP-responsive element-binding protein: A key role in glucose-mediated pancreatic beta-cell survival. Diabetes 2006, 55, 2220–2230.
  91. Hussain, M.A.; Porras, D.L.; Rowe, M.H.; West, J.R.; Song, W.J.; Schreiber, W.E.; Wondisford, F.E. Increased pancreatic beta-cell proliferation mediated by CREB binding protein gene activation. Mol. Cell. Biol. 2006, 26, 7747–7759.
  92. Piera-Velazquez, S.; Hawkins, D.F.; Whitecavage, M.K.; Colter, D.C.; Stokes, D.G.; Jimenez, S.A. Regulation of the human SOX9 promoter by Sp1 and CREB. Exp. Cell Res. 2007, 313, 1069–1079.
  93. Le Belle, J.E.; Orozco, N.M.; Paucar, A.A.; Saxe, J.P.; Mottahedeh, J.; Pyle, A.D.; Wu, H.; Kornblum, H.I. Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner. Cell Stem Cell 2011, 8, 59–71.
  94. Funato, Y.; Michiue, T.; Asashima, M.; Miki, H. The thioredoxin-related redox-regulating protein nucleoredoxin inhibits Wnt-beta-catenin signalling through dishevelled. Nat. Cell Biol. 2006, 8, 501–508.
  95. Fruhbeck, G. Intracellular signalling pathways activated by leptin. Biochem. J. 2006, 393, 7–20.
  96. Lee, Y.H.; Magkos, F.; Mantzoros, C.S.; Kang, E.S. Effects of leptin and adiponectin on pancreatic beta-cell function. Metabolism 2011, 60, 1664–1672.
  97. Kulkarni, R.N.; Wang, Z.L.; Wang, R.M.; Hurley, J.D.; Smith, D.M.; Ghatei, M.A.; Withers, D.J.; Gardiner, J.V.; Bailey, C.J.; Bloom, S.R. Leptin rapidly suppresses insulin release from insulinoma cells, rat and human islets and, in vivo, in mice. J. Clin. Investig. 1997, 100, 2729–2736.
  98. Kieffer, T.J.; Heller, R.S.; Leech, C.A.; Holz, G.G.; Habener, J.F. Leptin suppression of insulin secretion by the activation of ATP-sensitive K+ channels in pancreatic beta-cells. Diabetes 1997, 46, 1087–1093.
  99. Kuehnen, P.; Laubner, K.; Raile, K.; Schofl, C.; Jakob, F.; Pilz, I.; Path, G.; Seufert, J. Protein phosphatase 1 (PP-1)-dependent inhibition of insulin secretion by leptin in INS-1 pancreatic beta-cells and human pancreatic islets. Endocrinology 2011, 152, 1800–1808.
  100. Sim, A.T.; Baldwin, M.L.; Rostas, J.A.; Holst, J.; Ludowyke, R.I. The role of serine/threonine protein phosphatases in exocytosis. Biochem. J. 2003, 373, 641–659.
  101. Staiger, K.; Stefan, N.; Staiger, H.; Brendel, M.D.; Brandhorst, D.; Bretzel, R.G.; Machicao, F.; Kellerer, M.; Stumvoll, M.; Fritsche, A.; et al. Adiponectin is functionally active in human islets but does not affect insulin secretory function or beta-cell lipoapoptosis. J. Clin. Endocrinol. Metab. 2005, 90, 6707–6713.
  102. Llanos, P.; Contreras-Ferrat, A.; Barrientos, G.; Valencia, M.; Mears, D.; Hidalgo, C. Glucose-Dependent Insulin Secretion in Pancreatic beta-Cell Islets from Male Rats Requires Ca2+ Release via ROS-Stimulated Ryanodine Receptors. PLoS ONE 2015, 10, e0129238.
  103. Jansson, D.; Ng, A.C.; Fu, A.; Depatie, C.; Al Azzabi, M.; Screaton, R.A. Glucose controls CREB activity in islet cells via regulated phosphorylation of TORC2. Proc. Natl. Acad. Sci. USA 2008, 105, 10161–10166.
  104. Bernal-Mizrachi, E.; Kulkarni, R.N.; Scott, D.K.; Mauvais-Jarvis, F.; Stewart, A.F.; Garcia-Ocana, A. Human beta-cell proliferation and intracellular signaling part 2: Still driving in the dark without a road map. Diabetes 2014, 63, 819–831.
  105. Sato, Y.; Endo, H.; Okuyama, H.; Takeda, T.; Iwahashi, H.; Imagawa, A.; Yamagata, K.; Shimomura, I.; Inoue, M. Cellular hypoxia of pancreatic beta-cells due to high levels of oxygen consumption for insulin secretion in vitro. J. Biol. Chem. 2011, 286, 12524–12532.
  106. Zhdanov, A.V.; Ward, M.W.; Prehn, J.H.; Papkovsky, D.B. Dynamics of intracellular oxygen in PC12 Cells upon stimulation of neurotransmission. J. Biol. Chem. 2008, 283, 5650–5661.
  107. O’Hagan, K.A.; Cocchiglia, S.; Zhdanov, A.V.; Tambuwala, M.M.; Cummins, E.P.; Monfared, M.; Agbor, T.A.; Garvey, J.F.; Papkovsky, D.B.; Taylor, C.T.; et al. PGC-1alpha is coupled to HIF-1alpha-dependent gene expression by increasing mitochondrial oxygen consumption in skeletal muscle cells. Proc. Natl. Acad. Sci. USA 2009, 106, 2188–2193.
  108. Gerber, P.A.; Rutter, G.A. The Role of Oxidative Stress and Hypoxia in Pancreatic Beta-Cell Dysfunction in Diabetes Mellitus. Antioxid. Redox Signal. 2017, 26, 501–518.
  109. Olsson, R.; Carlsson, P.O. A low-oxygenated subpopulation of pancreatic islets constitutes a functional reserve of endocrine cells. Diabetes 2011, 60, 2068–2075.
  110. Ashcroft, F.M.; Harrison, D.E.; Ashcroft, S.J. Glucose induces closure of single potassium channels in isolated rat pancreatic beta-cells. Nature 1984, 312, 446–448.
  111. Yasui, S.; Mawatari, K.; Morizumi, R.; Furukawa, H.; Shimohata, T.; Harada, N.; Takahashi, A.; Nakaya, Y. Hydrogen peroxide inhibits insulin-induced ATP-sensitive potassium channel activation independent of insulin signaling pathway in cultured vascular smooth muscle cells. J. Med. Investig. 2012, 59, 36–44.
  112. Sakaguchi, R.; Mori, Y. Transient receptor potential (TRP) channels: Biosensors for redox environmental stimuli and cellular status. Free Radic. Biol. Med. 2020, 146, 36–44.
  113. Finol-Urdaneta, R.K.; Remedi, M.S.; Raasch, W.; Becker, S.; Clark, R.B.; Struver, N.; Pavlov, E.; Nichols, C.G.; French, R.J.; Terlau, H. Block of Kv1.7 potassium currents increases glucose-stimulated insulin secretion. EMBO Mol. Med. 2012, 4, 424–434.
  114. MacDonald, P.E.; Salapatek, A.M.; Wheeler, M.B. Temperature and redox state dependence of native Kv2.1 currents in rat pancreatic beta-cells. J. Physiol. 2003, 546, 647–653.
  115. Mittal, M.; Gu, X.Q.; Pak, O.; Pamenter, M.E.; Haag, D.; Fuchs, D.B.; Schermuly, R.T.; Ghofrani, H.A.; Brandes, R.P.; Seeger, W.; et al. Hypoxia induces Kv channel current inhibition by increased NADPH oxidase-derived reactive oxygen species. Free Radic. Biol. Med. 2012, 52, 1033–1042.
  116. Gerst, J.E. SNARE regulators: Matchmakers and matchbreakers. Biochim Biophys Acta 2003, 1641, 99–110.
  117. Ivarsson, R.; Quintens, R.; Dejonghe, S.; Tsukamoto, K.; Renstrom, E.; Schuit, F.C. Redox control of exocytosis: Regulatory role of NADPH, thioredoxin, and glutaredoxin. Diabetes 2005, 54, 2132–2142.
  118. Reinbothe, T.M.; Ivarsson, R.; Li, D.Q.; Niazi, O.; Jing, X.; Zhang, E.; Stenson, L.; Bryborn, U.; Renstrom, E. Glutaredoxin-1 mediates NADPH-dependent stimulation of calcium-dependent insulin secretion. Mol. Endocrinol. 2009, 23, 893–900.
  119. Ferdaoussi, M.; Dai, X.; Jensen, M.V.; Wang, R.; Peterson, B.S.; Huang, C.; Ilkayeva, O.; Smith, N.; Miller, N.; Hajmrle, C.; et al. Isocitrate-to-SENP1 signaling amplifies insulin secretion and rescues dysfunctional beta cells. J. Clin. Investig. 2015, 125, 3847–3860.
  120. Xu, Z.; Lam, L.S.; Lam, L.H.; Chau, S.F.; Ng, T.B.; Au, S.W. Molecular basis of the redox regulation of SUMO proteases: A protective mechanism of intermolecular disulfide linkage against irreversible sulfhydryl oxidation. FASEB J. 2008, 22, 127–137.
  121. Ferdaoussi, M.; MacDonald, P.E. Toward Connecting Metabolism to the Exocytotic Site. Trends Cell Biol. 2017, 27, 163–171.
  122. Lorenzen, I.; Eble, J.A.; Hanschmann, E.M. Thiol switches in membrane proteins - Extracellular redox regulation in cell biology. Biol. Chem. 2020.
  123. Lundberg, M.; Fernandes, A.P.; Kumar, S.; Holmgren, A. Cellular and plasma levels of human glutaredoxin 1 and 2 detected by sensitive ELISA systems. Biochem. Biophys. Res. Commun. 2004, 319, 801–809.
  124. Xiong, B.; Jha, V.; Min, J.K.; Cho, J. Protein disulfide isomerase in cardiovascular disease. Exp. Mol. Med. 2020, 52, 390–399.
  125. Mullen, L.; Hanschmann, E.M.; Lillig, C.H.; Herzenberg, L.A.; Ghezzi, P. Cysteine Oxidation Targets Peroxiredoxins 1 and 2 for Exosomal Release through a Novel Mechanism of Redox-Dependent Secretion. Mol. Med. 2015, 21, 98–108.
  126. Hanschmann, E.M.; Petry, S.F.; Eitner, S.; Maresch, C.C.; Lingwal, N.; Lillig, C.H.; Linn, T. Paracrine regulation and improvement of β-cell function by thioredoxin. Redox Biol. 2020, 34, 101570.
  127. Jikimoto, T.; Nishikubo, Y.; Koshiba, M.; Kanagawa, S.; Morinobu, S.; Morinobu, A.; Saura, R.; Mizuno, K.; Kondo, S.; Toyokuni, S.; et al. Thioredoxin as a biomarker for oxidative stress in patients with rheumatoid arthritis. Mol. Immunol. 2002, 38, 765–772.
  128. Kakisaka, Y.; Nakashima, T.; Sumida, Y.; Yoh, T.; Nakamura, H.; Yodoi, J.; Senmaru, H. Elevation of serum thioredoxin levels in patients with type 2 diabetes. Horm. Metab. Res. 2002, 34, 160–164.
  129. Asami, K.; Inagaki, A.; Imura, T.; Sekiguchi, S.; Fujimori, K.; Masutani, H.; Yodoi, J.; Satomi, S.; Ohuchi, N.; Goto, M. Thioredoxin-1 attenuates early graft loss after intraportal islet transplantation in mice. PLoS ONE 2013, 8, e70259.
  130. Willems, S.H.; Tape, C.J.; Stanley, P.L.; Taylor, N.A.; Mills, I.G.; Neal, D.E.; McCafferty, J.; Murphy, G. Thiol isomerases negatively regulate the cellular shedding activity of ADAM17. Biochem. J. 2010, 428, 439–450.
  131. Bass, R.; Edwards, D.R. ADAMs and protein disulfide isomerase: The key to regulated cell-surface protein ectodomain shedding? Biochem. J. 2010, 428, e3–e5.
  132. Düsterhöft, S.; Jung, S.; Hung, C.W.; Tholey, A.; Sönnichsen, F.D.; Grötzinger, J.; Lorenzen, I. Membrane-proximal domain of a disintegrin and metalloprotease-17 represents the putative molecular switch of its shedding activity operated by protein-disulfide isomerase. J. Am. Chem. Soc. 2013, 135, 5776–5781.
  133. Pedersen, K.B.; Chodavarapu, H.; Porretta, C.; Robinson, L.K.; Lazartigues, E. Dynamics of ADAM17-Mediated Shedding of ACE2 Applied to Pancreatic Islets of Male db/db Mice. Endocrinology 2015, 156, 4411–4425.
  134. Chhabra, K.H.; Chodavarapu, H.; Lazartigues, E. Angiotensin converting enzyme 2: A new important player in the regulation of glycemia. IUBMB Life 2013, 65, 731–738.
  135. Bergerhausen, L.; Grosche, J.; Meißner, J.; Hecker, C.; Caliandro, M.F.; Westerhausen, C.; Kamenac, A.; Rezaei, M.; Mörgelin, M.; Poschmann, G.; et al. Extracellular Redox Regulation of α7β Integrin-Mediated Cell Migration Is Signaled via a Dominant Thiol-Switch. Antioxidants 2020, 9, 227.
  136. Passam, F.; Chiu, J.; Ju, L.; Pijning, A.; Jahan, Z.; Mor-Cohen, R.; Yeheskel, A.; Kolšek, K.; Thärichen, L.; Aponte-Santamaría, C.; et al. Mechano-redox control of integrin de-adhesion. eLife 2018, 7.
  137. Townsend, S.E.; Gannon, M. Extracellular Matrix-Associated Factors Play Critical Roles in Regulating Pancreatic β-Cell Proliferation and Survival. Endocrinology 2019, 160, 1885–1894.
  138. Hynes, R.O. Integrins: Bidirectional, allosteric signaling machines. Cell 2002, 110, 673–687.
  139. Alam, N.; Goel, H.L.; Zarif, M.J.; Butterfield, J.E.; Perkins, H.M.; Sansoucy, B.G.; Sawyer, T.K.; Languino, L.R. The integrin-growth factor receptor duet. J. Cell. Physiol. 2007, 213, 649–653.
  140. Xu, S.Z.; Sukumar, P.; Zeng, F.; Li, J.; Jairaman, A.; English, A.; Naylor, J.; Ciurtin, C.; Majeed, Y.; Milligan, C.J.; et al. TRPC channel activation by extracellular thioredoxin. Nature 2008, 451, 69–72.
  141. Islam, M.S. Molecular Regulations and Functions of the Transient Receptor Potential Channels of the Islets of Langerhans and Insulinoma Cells. Cells 2020, 9, 685.
More
Video Production Service