Publication Cover
Redox Report
Communications in Free Radical Research
Volume 23, 2018 - Issue 1
2,532
Views
38
CrossRef citations to date
0
Altmetric
Research Articles

Renal mitochondrial oxidative stress is enhanced by the reduction of Sirt3 activity, in Zucker diabetic fatty rats

, , , ORCID Icon, &

References

  • Coresh J, Astor BC, Greene T, et al. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis. 2003;41:1–12. doi: 10.1053/ajkd.2003.50007
  • Hsu CY, Iribarren C, McCulloch CE, et al. Risk factors for end-stage renal disease: 25-year follow-up. Arch Intern Med. 2009;169:342–350. doi: 10.1001/archinternmed.2008.605
  • Chistiakov DA, Sobenin IA, Revin VV, et al. Mitochondrial aging and age-related dysfunction of mitochondria. Biomed Res Int. 2014;238463.
  • Kitada M, Kume S, Imaizumi N, et al. Resveratrol improves oxidative stress and protects against diabetic nephropathy through normalization of Mn-SOD dysfunction in AMPK/SIRT1-independent pathway. Diabetes. 2011;60:634–643. doi: 10.2337/db10-0386
  • Kitada M, Koya D, Sugimoto T, et al. Translocation of glomerular p47phox and p67phox by protein kinase C-beta activation is required for oxidative stress in diabetic nephropathy. Diabetes. 2003;52:2603–2614. doi: 10.2337/diabetes.52.10.2603
  • Koya D, Hayashi K, Kitada M, et al. Effects of antioxidants in diabetes-induced oxidative stress in the glomeruli of diabetic rats. J Am Soc Nephrol. 2003;14:S250–S253. doi: 10.1097/01.ASN.0000077412.07578.44
  • Guarente L, Franklin H. Epstein lecture: sirtuins, aging, and medicine. New Engl J Med. 2011;364:2235–2244. doi: 10.1056/NEJMra1100831
  • Someya S, Yu W, Hallows WC, et al. Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell. 2010;143:802–812. doi: 10.1016/j.cell.2010.10.002
  • Tao R, Vassilopoulos A, Parisiadou L, et al. Regulation of MnSOD enzymatic activity by Sirt3 connects the mitochondrial acetylome signaling networks to aging and carcinogenesis. Antioxid Redox Signal. 2014;20:1646–1654. doi: 10.1089/ars.2013.5482
  • Qiu X, Brown K, Hirschey MD, et al. Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab. 2010;12:662–667. doi: 10.1016/j.cmet.2010.11.015
  • Zhang L, Chen CL, Kang PT, et al. Differential protein acetylation assists import of excess SOD2 into mitochondria and mediates SOD2 aggregation associated with cardiac hypertrophy in the murine SOD2-tg heart. Free Radic Biol Med. 2017;108:595–609. doi: 10.1016/j.freeradbiomed.2017.04.022
  • Morigi M, Perico L, Rota C, et al. Sirtuin 3-dependent mitochondrial dynamic improvements protect against acute kidney injury. J Clin Invest. 2015;125:715–726. doi: 10.1172/JCI77632
  • Kitada M, Kume S, Takeda-Watanabe A, et al. Sirtuins and renal diseases: relationship with aging and diabetic nephropathy. Clin Sci (Lond). 2013;124:153–164. doi: 10.1042/CS20120190
  • Braidy N, Guillemin GJ, Mansour H, et al. Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in wistar rats. PLoS One. 2011;6:e19194. doi: 10.1371/journal.pone.0019194
  • Aksoy P, White TA, Thompson M, et al. Regulation of intracellular levels of NAD: a novel role for CD38. Biochem Biophys Res Commun. 2006;345:1386–1392. doi: 10.1016/j.bbrc.2006.05.042
  • Camacho-Pereira J, Tarrago MG, Chini CCS, et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab. 2016;23:1127–1139. doi: 10.1016/j.cmet.2016.05.006
  • Kitada M, Ogura Y, Suzuki T, et al. A very-low-protein diet ameliorates advanced diabetic nephropathy through autophagy induction by suppression of the mTORC1 pathway in Wistar fatty rats, an animal model of type 2 diabetes and obesity. Diabetologia. 2016;59:1307–1317.
  • Nagai T, Kanasaki M, Srivastava SP, et al. N-acetyl-seryl-aspartyl-lysyl-proline inhibits diabetes-associated kidney fibrosis and endothelial-mesenchymal transition. Biomed Res Int. 2014;2014: 696475.
  • Ozdemir BC, Pentcheva-Hoang T, Carstens JL, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2015;28:831–833. doi: 10.1016/j.ccell.2015.11.002
  • Kume S, Uzu T, Horiike K, et al. Calorie restriction enhances cell adaptation to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged kidney. J Clin Invest. 2010;120:1043–1055. doi: 10.1172/JCI41376
  • Bonkowski MS, Sinclair DA. Slowing ageing by design: the rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol. 2016;17:679–690. doi: 10.1038/nrm.2016.93
  • Hebert AS, Dittenhafer-Reed KE, Yu W, et al. Calorie restriction and SIRT3 trigger global reprogramming of the mitochondrial protein acetylome. Moll Cell. 2013;49:186–199. doi: 10.1016/j.molcel.2012.10.024
  • Yu W, Dittenhafer-Reed KE, Denu JM. SIRT3 protein deacetylates isocitrate dehydrogenase 2 (IDH2) and regulates mitochondrial redox status. J Biol Chem. 2012;287:14078–14086. doi: 10.1074/jbc.M112.355206
  • Zhu XH, Lu M, Lee BY, et al. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc Natl Acad Sci U S A. 2015;112:2876–2881. doi: 10.1073/pnas.1417921112
  • Gomes AP, Price NL, Ling AJ, et al. Declining NAD(+) induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell. 2013;155:1624–1638. doi: 10.1016/j.cell.2013.11.037
  • Massudi H, Grant R, Braidy N, et al. Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PLoS One. 2012;7:e42357. doi: 10.1371/journal.pone.0042357
  • Liang M, Chini EN, Cheng J, et al. Synthesis of NAADP and cADPR in mitochondria. Arch Biochem Biophys. 1999;371:317–325. doi: 10.1006/abbi.1999.1463
  • Sun L, Adebanjo OA, Koval A, et al. A novel mechanism for coupling cellular intermediary metabolism to cytosolic Ca2+ signaling via CD38/ADP-ribosyl cyclase, a putative intracellular NAD+ sensor. FASEB J. 2002;16:302–314. doi: 10.1096/fj.01-0705com
  • Yamada M, Mizuguchi M, Otsuka N, et al. Ultrastructural localization of CD38 immunoreactivity in rat brain. Brain Res. 1997;756:52–60. doi: 10.1016/S0006-8993(97)00117-0
  • Escande C, Nin V, Price NL, et al. Flavonoid apigenin is an inhibitor of the NAD+ ase CD38: implications for cellular NAD+ metabolism, protein acetylation, and treatment of metabolic syndrome. Diabetes. 2013;62:1084–1093. doi: 10.2337/db12-1139
  • Barata H, Thompson M, Zielinska W, et al. The role of cyclic-ADP-ribose-signaling pathway in oxytocin-induced Ca2+ transients in human myometrium cells. Endocrinology. 2004;145:881–889. doi: 10.1210/en.2003-0774
  • Ma Y, Wu D, Ding X, et al. CD38 plays key roles in both antioxidation and cell survival of H2O2-treated primary rodent astrocytes. Int J Physiol Pathophysiol Pharmacol. 2014;6:102–108.