Sirtuins and Renal Oxidative Stress
Abstract
:1. Kidney and Oxidative Stress
2. Regulation of Oxidative Stress
2.1. Oxidative Stress Mechanisms
2.2. Antioxidant Defense Mechanisms
2.3. Hormesis and Oxidative Stress
3. Renal Oxidative Stress and Anti-Aging Therapy
4. Kidney and Sirtuins
5. Sirtuins and Renal Oxidative Stress
5.1. Sirt1
5.1.1. Sirt1 and NF-ĸB
5.1.2. Sirt1 and p53
5.1.3. Sirt1 and eNOS
5.1.4. Sirt1 and p66shc
5.1.5. Sirt1 and PGC-1α
5.1.6. Sirt1 and AMPK
5.1.7. Sirt1 and FoxO
5.1.8. Sirt1 and HIF-1α
5.2. Sirt2
5.3. Sirt3
5.4. Sirt4
5.5. Sirt5
5.6. Sirt6
5.7. Sirt7
5.8. Sirtuin Activators and Their Effects
6. Considerations and Conclusions for Future Research
Author Contributions
Funding
Conflicts of Interest
References
- Hill, N.R.; Fatoba, S.T.; Oke, J.L.; Hirst, J.A.; O’Callaghan, C.A.; Lasserson, D.S.; Hobbs, F.D. Global Prevalence of Chronic Kidney Disease—A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0158765. [Google Scholar] [CrossRef] [PubMed]
- Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef]
- Qian, Q. Inflammation: A Key Contributor to the Genesis and Progression of Chronic Kidney Disease. Contrib. Nephrol. 2017, 191, 72–83. [Google Scholar] [CrossRef] [PubMed]
- Nangaku, M. Chronic hypoxia and tubulointerstitial injury: A final common pathway to end-stage renal failure. J. Am. Soc. Nephrol. JASN 2006, 17, 17–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mimura, I.; Nangaku, M. The suffocating kidney: Tubulointerstitial hypoxia in end-stage renal disease. Nat. Rev. Nephrol. 2010, 6, 667–678. [Google Scholar] [CrossRef]
- Ow, C.P.C.; Ngo, J.P.; Ullah, M.M.; Hilliard, L.M.; Evans, R.G. Renal hypoxia in kidney disease: Cause or consequence? Acta Physiol. 2018, 222, e12999. [Google Scholar] [CrossRef]
- Poulianiti, K.P.; Kaltsatou, A.; Mitrou, G.I.; Jamurtas, A.Z.; Koutedakis, Y.; Maridaki, M.; Stefanidis, I.; Sakkas, G.K.; Karatzaferi, C. Systemic Redox Imbalance in Chronic Kidney Disease: A Systematic Review. Oxid. Med. Cell Longev. 2016, 2016, 8598253. [Google Scholar] [CrossRef] [Green Version]
- Liakopoulos, V.; Roumeliotis, S.; Gorny, X.; Eleftheriadis, T.; Mertens, P.R. Oxidative Stress in Patients Undergoing Peritoneal Dialysis: A Current Review of the Literature. Oxid. Med. Cell Longev. 2017, 2017, 3494867. [Google Scholar] [CrossRef] [Green Version]
- Kooman, J.P.; van der Sande, F.M.; Leunissen, K.M. Kidney disease and aging: A reciprocal relation. Exp. Gerontol. 2017, 87, 156–159. [Google Scholar] [CrossRef]
- Kovesdy, C.P.; Furth, S.; Zoccali, C.; World Kidney Day Steering, C. Obesity and kidney disease: Hidden consequences of the epidemic. Indian J. Nephrol. 2017, 27, 85–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity: Implications for metabolic syndrome, diabetes, hypertension, dyslipidemia, atherosclerosis, and cancer. Obes. Res. Clin. Pract. 2013, 7, e330–e341. [Google Scholar] [CrossRef]
- Diabetes and Kidney Disease: Role of Oxidative Stress. Antioxid. Redox Signal. 2016, 25, 657–684. [CrossRef] [PubMed] [Green Version]
- Tsujimoto, T.; Sairenchi, T.; Iso, H.; Irie, F.; Yamagishi, K.; Watanabe, H.; Tanaka, K.; Muto, T.; Ota, H. The dose-response relationship between body mass index and the risk of incident stage ≥3 chronic kidney disease in a general japanese population: The Ibaraki prefectural health study (IPHS). J. Epidemiol. 2014, 24, 444–451. [Google Scholar] [CrossRef] [Green Version]
- Nangaku, M.; Hirakawa, Y.; Mimura, I.; Inagi, R.; Tanaka, T. Epigenetic Changes in the Acute Kidney Injury-to-Chronic Kidney Disease Transition. Nephron 2017, 137, 256–259. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, S.; Tanaka, T.; Nangaku, M. Hypoxia as a key player in the AKI-to-CKD transition. Am. J. Physiol. Ren. Physiol. 2014, 307, F1187–F1195. [Google Scholar] [CrossRef] [Green Version]
- Shu, S.; Wang, Y.; Zheng, M.; Liu, Z.; Cai, J.; Tang, C.; Dong, Z. Hypoxia and Hypoxia-Inducible Factors in Kidney Injury and Repair. Cells 2019, 8, 207. [Google Scholar] [CrossRef] [Green Version]
- Cristóbal-García, M.; García-Arroyo, F.E.; Tapia, E.; Osorio, H.; Arellano-Buendía, A.S.; Madero, M.; Rodríguez-Iturbe, B.; Pedraza-Chaverrí, J.; Correa, F.; Zazueta, C.; et al. Renal Oxidative Stress Induced by Long-Term Hyperuricemia Alters Mitochondrial Function and Maintains Systemic Hypertension. Oxid. Med. Cell Longev. 2015, 2015, 535686. [Google Scholar] [CrossRef] [Green Version]
- Palm, F.; Nordquist, L. Renal oxidative stress, oxygenation, and hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 301, R1229–R1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salisbury, D.; Bronas, U. Reactive Oxygen and Nitrogen Species: Impact on Endothelial Dysfunction. Nurs. Res. 2015, 64, 53–66. [Google Scholar] [CrossRef] [PubMed]
- Jones, D.P.; Sies, H. The Redox Code. Antioxid. Redox Signal. 2015, 23, 734–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Powers, S.K.; Ji, L.L.; Kavazis, A.N.; Jackson, M.J. Reactive oxygen species: Impact on skeletal muscle. Compr. Physiol. 2011, 1, 941–969. [Google Scholar] [CrossRef] [Green Version]
- Bae, Y.S.; Oh, H.; Rhee, S.G.; Yoo, Y.D. Regulation of reactive oxygen species generation in cell signaling. Mol. Cells 2011, 32, 491–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pham-Huy, L.A.; He, H.; Pham-Huy, C. Free radicals, antioxidants in disease and health. Int. J. Biomed. Sci. IJBS 2008, 4, 89–96. [Google Scholar] [PubMed]
- Halliwell, B. Biochemistry of oxidative stress. Biochem. Soc. Trans. 2007, 35, 1147–1150. [Google Scholar] [CrossRef] [PubMed]
- Weidinger, A.; Kozlov, A.V. Biological Activities of Reactive Oxygen and Nitrogen Species: Oxidative Stress versus Signal Transduction. Biomolecules 2015, 5, 472–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Genestra, M. Oxyl radicals, redox-sensitive signalling cascades and antioxidants. Cell. Signal. 2007, 19, 1807–1819. [Google Scholar] [CrossRef] [PubMed]
- Kulcharyk, P.A.; Heinecke, J.W. Hypochlorous acid produced by the myeloperoxidase system of human phagocytes induces covalent cross-links between DNA and protein. Biochemistry 2001, 40, 3648–3656. [Google Scholar] [CrossRef] [PubMed]
- Adams, L.; Franco, M.C.; Estevez, A.G. Reactive nitrogen species in cellular signaling. Exp. Biol. Med. 2015, 240, 711–717. [Google Scholar] [CrossRef] [Green Version]
- Mikkelsen, R.B.; Wardman, P. Biological chemistry of reactive oxygen and nitrogen and radiation-induced signal transduction mechanisms. Oncogene 2003, 22, 5734–5754. [Google Scholar] [CrossRef] [Green Version]
- Ridnour, L.A.; Thomas, D.D.; Mancardi, D.; Espey, M.G.; Miranda, K.M.; Paolocci, N.; Feelisch, M.; Fukuto, J.; Wink, D.A. The chemistry of nitrosative stress induced by nitric oxide and reactive nitrogen oxide species. Putting perspective on stressful biological situations. Biol. Chem. 2004, 385, 1–10. [Google Scholar] [CrossRef]
- Phaniendra, A.; Jestadi, D.B.; Periyasamy, L. Free radicals: Properties, sources, targets, and their implication in various diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [Green Version]
- Young, I.S.; Woodside, J.V. Antioxidants in health and disease. J. Clin. Pathol. 2001, 54, 176–186. [Google Scholar] [CrossRef] [Green Version]
- Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [Green Version]
- Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campbell, N.K.; Fitzgerald, H.K.; Dunne, A. Regulation of inflammation by the antioxidant haem oxygenase 1. Nat. Rev. Immunol. 2021, 1–15. [Google Scholar] [CrossRef]
- Morimoto, R.I. Proteotoxic stress and inducible chaperone networks in neurodegenerative disease and aging. Genes Dev. 2008, 22, 1427–1438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cornelius, C.; Perrotta, R.; Graziano, A.; Calabrese, E.J.; Calabrese, V. Stress responses, vitagenes and hormesis as critical determinants in aging and longevity: Mitochondria as a “chi”. Immun. Ageing I A 2013, 10, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Calabrese, E.J.; Mattson, M.P.; Calabrese, V. Resveratrol commonly displays hormesis: Occurrence and biomedical significance. Hum. Exp. Toxicol. 2010, 29, 980–1015. [Google Scholar] [CrossRef] [PubMed]
- Di Paola, R.; Impellizzeri, D.; Salinaro, A.T.; Mazzon, E.; Bellia, F.; Cavallaro, M.; Cornelius, C.; Vecchio, G.; Calabrese, V.; Rizzarelli, E.; et al. Administration of carnosine in the treatment of acute spinal cord injury. Biochem. Pharmacol. 2011, 82, 1478–1489. [Google Scholar] [CrossRef]
- Pennisi, G.; Cornelius, C.; Cavallaro, M.M.; Salinaro, A.T.; Cambria, M.T.; Pennisi, M.; Bella, R.; Milone, P.; Ventimiglia, B.; Migliore, M.R.; et al. Redox regulation of cellular stress response in multiple sclerosis. Biochem. Pharmacol. 2011, 82, 1490–1499. [Google Scholar] [CrossRef] [Green Version]
- Scapagnini, G.; Caruso, C.; Calabrese, V. Therapeutic potential of dietary polyphenols against brain ageing and neurodegenerative disorders. Adv. Exp. Med. Biol. 2010, 698, 27–35. [Google Scholar] [CrossRef] [PubMed]
- Bellia, F.; Vecchio, G.; Cuzzocrea, S.; Calabrese, V.; Rizzarelli, E. Neuroprotective features of carnosine in oxidative driven diseases. Mol. Asp. Med. 2011, 32, 258–266. [Google Scholar] [CrossRef]
- Kobayashi, A.; Kang, M.I.; Okawa, H.; Ohtsuji, M.; Zenke, Y.; Chiba, T.; Igarashi, K.; Yamamoto, M. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell Biol. 2004, 24, 7130–7139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Forman, H.J. Reexamination of the electrophile response element sequences and context reveals a lack of consensus in gene function. Biochim. Biophys. Acta 2010, 1799, 496–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- García Trejo, E.M.Á.; Arellano Buendía, A.S.; Sánchez Reyes, O.; García Arroyo, F.E.; Arguello García, R.; Loredo Mendoza, M.L.; Tapia, E.; Sánchez Lozada, L.G.; Osorio Alonso, H. The Beneficial Effects of Allicin in Chronic Kidney Disease Are Comparable to Losartan. Int. J. Mol. Sci. 2017, 18, 1980. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.Q.; Kosten, T.R.; Zhang, X.Y. Free radicals, antioxidant defense systems, and schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2013, 46, 200–206. [Google Scholar] [CrossRef]
- Massy, Z.A.; Nguyen-Khoa, T. Oxidative stress and chronic renal failure: Markers and management. J. Nephrol. 2002, 15, 336–341. [Google Scholar]
- Small, D.M.; Coombes, J.S.; Bennett, N.; Johnson, D.W.; Gobe, G.C. Oxidative stress, anti-oxidant therapies and chronic kidney disease. Nephrology 2012, 17, 311–321. [Google Scholar] [CrossRef]
- Dounousi, E.; Papavasiliou, E.; Makedou, A.; Ioannou, K.; Katopodis, K.P.; Tselepis, A.; Siamopoulos, K.C.; Tsakiris, D. Oxidative stress is progressively enhanced with advancing stages of CKD. Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 2006, 48, 752–760. [Google Scholar] [CrossRef] [Green Version]
- Nistala, R.; Whaley-Connell, A.; Sowers, J.R. Redox control of renal function and hypertension. Antioxid. Redox Signal. 2008, 10, 2047–2089. [Google Scholar] [CrossRef] [Green Version]
- Pisoschi, A.M.; Pop, A. The role of antioxidants in the chemistry of oxidative stress: A review. Eur. J. Med. Chem. 2015, 97, 55–74. [Google Scholar] [CrossRef]
- Siracusa, R.; Scuto, M.; Fusco, R.; Trovato, A.; Ontario, M.L.; Crea, R.; Di Paola, R.; Cuzzocrea, S.; Calabrese, V. Anti-inflammatory and Anti-oxidant Activity of Hidrox(®) in Rotenone-Induced Parkinson’s Disease in Mice. Antioxidants 2020, 9, 824. [Google Scholar] [CrossRef] [PubMed]
- Calabrese, V.; Cornelius, C.; Dinkova-Kostova, A.T.; Calabrese, E.J.; Mattson, M.P. Cellular stress responses, the hormesis paradigm, and vitagenes: Novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid. Redox Signal. 2010, 13, 1763–1811. [Google Scholar] [CrossRef]
- Calabrese, E.J.; Iavicoli, I.; Calabrese, V. Hormesis: Why it is important to biogerontologists. Biogerontology 2012, 13, 215–235. [Google Scholar] [CrossRef]
- Stebbing, A.R. Hormesis--the stimulation of growth by low levels of inhibitors. Sci. Total Environ. 1982, 22, 213–234. [Google Scholar] [CrossRef]
- Sachdev, S.; Davies, K.J. Production, detection, and adaptive responses to free radicals in exercise. Free Radic. Biol. Med. 2008, 44, 215–223. [Google Scholar] [CrossRef] [PubMed]
- Shan, X.; Chi, L.; Ke, Y.; Luo, C.; Qian, S.; Gozal, D.; Liu, R. Manganese superoxide dismutase protects mouse cortical neurons from chronic intermittent hypoxia-mediated oxidative damage. Neurobiol. Dis. 2007, 28, 206–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keller, J.N.; Kindy, M.S.; Holtsberg, F.W.; St Clair, D.K.; Yen, H.C.; Germeyer, A.; Steiner, S.M.; Bruce-Keller, A.J.; Hutchins, J.B.; Mattson, M.P. Mitochondrial manganese superoxide dismutase prevents neural apoptosis and reduces ischemic brain injury: Suppression of peroxynitrite production, lipid peroxidation, and mitochondrial dysfunction. J. Neurosci. Off. J. Soc. Neurosci. 1998, 18, 687–697. [Google Scholar] [CrossRef] [Green Version]
- Ekdahl, C.T.; Kokaia, Z.; Lindvall, O. Brain inflammation and adult neurogenesis: The dual role of microglia. Neuroscience 2009, 158, 1021–1029. [Google Scholar] [CrossRef]
- Jiang, F.; Zhang, Z.G.; Katakowski, M.; Robin, A.M.; Faber, M.; Zhang, F.; Chopp, M. Angiogenesis induced by photodynamic therapy in normal rat brains. Photochem. Photobiol. 2004, 79, 494–498. [Google Scholar] [CrossRef]
- Maulik, N.; Das, D.K. Redox signaling in vascular angiogenesis. Free Radic. Biol. Med. 2002, 33, 1047–1060. [Google Scholar] [CrossRef]
- Calabrese, V.; Cornelius, C.; Cuzzocrea, S.; Iavicoli, I.; Rizzarelli, E.; Calabrese, E.J. Hormesis, cellular stress response and vitagenes as critical determinants in aging and longevity. Mol. Asp. Med. 2011, 32, 279–304. [Google Scholar] [CrossRef] [PubMed]
- Calabrese, E.J.; Iavicoli, I.; Calabrese, V. Hormesis: Its impact on medicine and health. Hum. Exp. Toxicol. 2013, 32, 120–152. [Google Scholar] [CrossRef] [PubMed]
- Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nitta, K.; Okada, K.; Yanai, M.; Takahashi, S. Aging and chronic kidney disease. Kidney Blood Press. Res. 2013, 38, 109–120. [Google Scholar] [CrossRef]
- Wang, J.C.; Bennett, M. Aging and atherosclerosis: Mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ. Res. 2012, 111, 245–259. [Google Scholar] [CrossRef] [Green Version]
- Walsh, M.E.; Shi, Y.; Van Remmen, H. The effects of dietary restriction on oxidative stress in rodents. Free Radic. Biol. Med. 2014, 66, 88–99. [Google Scholar] [CrossRef] [Green Version]
- Fontana, L.; Partridge, L.; Longo, V.D. Extending healthy life span—From yeast to humans. Science 2010, 328, 321–326. [Google Scholar] [CrossRef] [Green Version]
- Lin, S.J.; Kaeberlein, M.; Andalis, A.A.; Sturtz, L.A.; Defossez, P.A.; Culotta, V.C.; Fink, G.R.; Guarente, L. Calorie restriction extends Saccharomyces cerevisiae lifespan by increasing respiration. Nature 2002, 418, 344–348. [Google Scholar] [CrossRef]
- Cohen, H.Y.; Miller, C.; Bitterman, K.J.; Wall, N.R.; Hekking, B.; Kessler, B.; Howitz, K.T.; Gorospe, M.; de Cabo, R.; Sinclair, D.A. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 2004, 305, 390–392. [Google Scholar] [CrossRef] [Green Version]
- Guarente, L. Calorie restriction and sirtuins revisited. Genes Dev. 2013, 27, 2072–2085. [Google Scholar] [CrossRef] [Green Version]
- Borradaile, N.M.; Pickering, J.G. Nicotinamide phosphoribosyltransferase imparts human endothelial cells with extended replicative lifespan and enhanced angiogenic capacity in a high glucose environment. Aging Cell 2009, 8, 100–112. [Google Scholar] [CrossRef]
- Jedrusik-Bode, M.; Studencka, M.; Smolka, C.; Baumann, T.; Schmidt, H.; Kampf, J.; Paap, F.; Martin, S.; Tazi, J.; Müller, K.M.; et al. The sirtuin SIRT6 regulates stress granule formation in C. elegans and mammals. J. Cell Sci. 2013, 126, 5166–5177. [Google Scholar] [CrossRef] [Green Version]
- Bao, X.; Wang, Y.; Li, X.; Li, X.M.; Liu, Z.; Yang, T.; Wong, C.F.; Zhang, J.; Hao, Q.; Li, X.D. Identification of ‘erasers’ for lysine crotonylated histone marks using a chemical proteomics approach. eLife 2014, 3. [Google Scholar] [CrossRef] [Green Version]
- Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kitada, M.; Ogura, Y.; Monno, I.; Koya, D. Sirtuins and Type 2 Diabetes: Role in Inflammation, Oxidative Stress, and Mitochondrial Function. Front. Endocrinol. 2019, 10, 187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Radak, Z.; Koltai, E.; Taylor, A.W.; Higuchi, M.; Kumagai, S.; Ohno, H.; Goto, S.; Boldogh, I. Redox-regulating sirtuins in aging, caloric restriction, and exercise. Free Radic. Biol. Med. 2013, 58, 87–97. [Google Scholar] [CrossRef] [PubMed]
- Merksamer, P.I.; Liu, Y.; He, W.; Hirschey, M.D.; Chen, D.; Verdin, E. The sirtuins, oxidative stress and aging: An emerging link. Aging 2013, 5, 144–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Webster, B.R.; Lu, Z.; Sack, M.N.; Scott, I. The role of sirtuins in modulating redox stressors. Free Radic. Biol. Med. 2012, 52, 281–290. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.T.; Wu, S.B.; Wei, Y.H. Roles of sirtuins in the regulation of antioxidant defense and bioenergetic function of mitochondria under oxidative stress. Free Radic. Res. 2014, 48, 1070–1084. [Google Scholar] [CrossRef]
- Bai, W.; Zhang, X. Nucleus or cytoplasm? The mysterious case of SIRT1’s subcellular localization. Cell Cycle 2016, 15, 3337–3338. [Google Scholar] [CrossRef] [Green Version]
- Yu, J.; Auwerx, J. Protein deacetylation by SIRT1: An emerging key post-translational modification in metabolic regulation. Pharmacol. Res. 2010, 62, 35–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imai, S.; Armstrong, C.M.; Kaeberlein, M.; Guarente, L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 2000, 403, 795–800. [Google Scholar] [CrossRef]
- Kitada, M.; Kume, S.; Kanasaki, K.; Takeda-Watanabe, A.; Koya, D. Sirtuins as possible drug targets in type 2 diabetes. Curr. Drug Targets 2013, 14, 622–636. [Google Scholar] [CrossRef]
- Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, L.; Xu, D.Y.; Sha, W.G.; Shen, L.; Lu, G.Y.; Yin, X.; Wang, M.J. High glucose induces renal tubular epithelial injury via Sirt1/NF-kappaB/microR-29/Keap1 signal pathway. J. Transl. Med. 2015, 13, 352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xue, F.; Huang, J.W.; Ding, P.Y.; Zang, H.G.; Kou, Z.J.; Li, T.; Fan, J.; Peng, Z.W.; Yan, W.J. Nrf2/antioxidant defense pathway is involved in the neuroprotective effects of Sirt1 against focal cerebral ischemia in rats after hyperbaric oxygen preconditioning. Behav. Brain Res. 2016, 309, 1–8. [Google Scholar] [CrossRef]
- Li, X.; Cai, W.; Lee, K.; Liu, B.; Deng, Y.; Chen, Y.; Zhang, X.; He, J.C.; Zhong, Y. Puerarin attenuates diabetic kidney injury through the suppression of NOX4 expression in podocytes. Sci. Rep. 2017, 7, 14603. [Google Scholar] [CrossRef]
- Schmid, H.; Boucherot, A.; Yasuda, Y.; Henger, A.; Brunner, B.; Eichinger, F.; Nitsche, A.; Kiss, E.; Bleich, M.; Grone, H.J.; et al. Modular activation of nuclear factor-kappaB transcriptional programs in human diabetic nephropathy. Diabetes 2006, 55, 2993–3003. [Google Scholar] [CrossRef] [Green Version]
- Liu, R.; Zhong, Y.; Li, X.; Chen, H.; Jim, B.; Zhou, M.M.; Chuang, P.Y.; He, J.C. Role of transcription factor acetylation in diabetic kidney disease. Diabetes 2014, 63, 2440–2453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andrade-Oliveira, V.; Foresto-Neto, O.; Watanabe, I.; Zatz, R.; Câmara, N.J.F.i.P. Inflammation in Renal Diseases: New and Old Players. Front. Pharmacol. 2019, 10, 1192. [Google Scholar] [CrossRef]
- Ong, A.L.C.; Ramasamy, T.S. Role of Sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res. Rev. 2018, 43, 64–80. [Google Scholar] [CrossRef] [PubMed]
- Kao, C.L.; Chen, L.K.; Chang, Y.L.; Yung, M.C.; Hsu, C.C.; Chen, Y.C.; Lo, W.L.; Chen, S.J.; Ku, H.H.; Hwang, S.J. Resveratrol protects human endothelium from H(2)O(2)-induced oxidative stress and senescence via SirT1 activation. J. Atheroscler. Thromb. 2010, 17, 970–979. [Google Scholar] [CrossRef] [Green Version]
- Ota, H.; Akishita, M.; Eto, M.; Iijima, K.; Kaneki, M.; Ouchi, Y. Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J. Mol. Cell. Cardiol. 2007, 43, 571–579. [Google Scholar] [CrossRef] [PubMed]
- Maillet, A.; Pervaiz, S. Redox regulation of p53, redox effectors regulated by p53: A subtle balance. Antioxid. Redox Signal. 2012, 16, 1285–1294. [Google Scholar] [CrossRef] [PubMed]
- Hussain, S.P.; Amstad, P.; He, P.; Robles, A.; Lupold, S.; Kaneko, I.; Ichimiya, M.; Sengupta, S.; Mechanic, L.; Okamura, S.; et al. p53-induced up-regulation of MnSOD and GPx but not catalase increases oxidative stress and apoptosis. Cancer Res. 2004, 64, 2350–2356. [Google Scholar] [CrossRef] [Green Version]
- Sablina, A.A.; Budanov, A.V.; Ilyinskaya, G.V.; Agapova, L.S.; Kravchenko, J.E.; Chumakov, P.M. The antioxidant function of the p53 tumor suppressor. Nat. Med. 2005, 11, 1306–1313. [Google Scholar] [CrossRef] [Green Version]
- Tan, M.; Li, S.; Swaroop, M.; Guan, K.; Oberley, L.W.; Sun, Y. Transcriptional Activation of the Human Glutathione Peroxidase Promoter by p53*. J. Biol. Chem. 1999, 274, 12061–12066. [Google Scholar] [CrossRef] [Green Version]
- Vaziri, H.; Dessain, S.K.; Ng Eaton, E.; Imai, S.I.; Frye, R.A.; Pandita, T.K.; Guarente, L.; Weinberg, R.A. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell 2001, 107, 149–159. [Google Scholar] [CrossRef] [Green Version]
- Tikoo, K.; Tripathi, D.N.; Kabra, D.G.; Sharma, V.; Gaikwad, A.B. Intermittent fasting prevents the progression of type I diabetic nephropathy in rats and changes the expression of Sir2 and p53. FEBS Lett. 2007, 581, 1071–1078. [Google Scholar] [CrossRef] [Green Version]
- Niranjan, T.; Murea, M.; Susztak, K. The pathogenic role of Notch activation in podocytes. Nephron. Exp. Nephrol. 2009, 111, e73–e79. [Google Scholar] [CrossRef] [Green Version]
- Donato, A.J.; Magerko, K.A.; Lawson, B.R.; Durrant, J.R.; Lesniewski, L.A.; Seals, D.R. SIRT-1 and vascular endothelial dysfunction with ageing in mice and humans. J. Physiol. 2011, 589, 4545–4554. [Google Scholar] [CrossRef] [PubMed]
- Mattagajasingh, I.; Kim, C.S.; Naqvi, A.; Yamamori, T.; Hoffman, T.A.; Jung, S.B.; DeRicco, J.; Kasuno, K.; Irani, K. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 2007, 104, 14855–14860. [Google Scholar] [CrossRef] [Green Version]
- Lempiäinen, J.; Finckenberg, P.; Mervaala, E.E.; Sankari, S.; Levijoki, J.; Mervaala, E.M. Caloric restriction ameliorates kidney ischaemia/reperfusion injury through PGC-1α-eNOS pathway and enhanced autophagy. Acta Physiol. 2013, 208, 410–421. [Google Scholar] [CrossRef] [PubMed]
- Song, P.; Yang, S.; Xiao, L.; Xu, X.; Tang, C.; Yang, Y.; Ma, M.; Zhu, J.; Liu, F.; Sun, L. PKCδ promotes high glucose induced renal tubular oxidative damage via regulating activation and translocation of p66Shc. Oxid. Med. Cell Longev. 2014, 2014, 746531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, S.; Kim, Y.R.; Vikram, A.; Naqvi, A.; Li, Q.; Kassan, M.; Kumar, V.; Bachschmid, M.M.; Jacobs, J.S.; Kumar, A.; et al. Sirtuin1-regulated lysine acetylation of p66Shc governs diabetes-induced vascular oxidative stress and endothelial dysfunction. Proc. Natl. Acad. Sci. USA 2017, 114, 1714–1719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nemoto, S.; Finkel, T. Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway. Science 2002, 295, 2450–2452. [Google Scholar] [CrossRef] [PubMed]
- Zhou, S.; Chen, H.Z.; Wan, Y.Z.; Zhang, Q.J.; Wei, Y.S.; Huang, S.; Liu, J.J.; Lu, Y.B.; Zhang, Z.Q.; Yang, R.F.; et al. Repression of P66Shc expression by SIRT1 contributes to the prevention of hyperglycemia-induced endothelial dysfunction. Circ. Res. 2011, 109, 639–648. [Google Scholar] [CrossRef]
- Chen, H.; Wan, Y.; Zhou, S.; Lu, Y.; Zhang, Z.; Zhang, R.; Chen, F.; Hao, D.; Zhao, X.; Guo, Z.; et al. Endothelium-specific SIRT1 overexpression inhibits hyperglycemia-induced upregulation of vascular cell senescence. Sci. China Life Sci. 2012, 55, 467–473. [Google Scholar] [CrossRef] [Green Version]
- Rota, M.; LeCapitaine, N.; Hosoda, T.; Boni, A.; De Angelis, A.; Padin-Iruegas, M.E.; Esposito, G.; Vitale, S.; Urbanek, K.; Casarsa, C.; et al. Diabetes promotes cardiac stem cell aging and heart failure, which are prevented by deletion of the p66shc gene. Circ. Res. 2006, 99, 42–52. [Google Scholar] [CrossRef] [Green Version]
- Menini, S.; Amadio, L.; Oddi, G.; Ricci, C.; Pesce, C.; Pugliese, F.; Giorgio, M.; Migliaccio, E.; Pelicci, P.; Iacobini, C.; et al. Erratum. Deletion of p66(Shc) Longevity Gene Protects Against Experimental Diabetic Glomerulopathy by Preventing Diabetes-Induced Oxidative Stress. Diabetes 2006;55:1642–1650. Diabetes 2018, 67, 165. [Google Scholar] [CrossRef] [Green Version]
- Wright, K.D.; Staruschenko, A.; Sorokin, A. Role of adaptor protein p66Shc in renal pathologies. Am. J. Physiol. Ren. Physiol. 2018, 314, F143–F153. [Google Scholar] [CrossRef]
- Gerhart-Hines, Z.; Rodgers, J.T.; Bare, O.; Lerin, C.; Kim, S.H.; Mostoslavsky, R.; Alt, F.W.; Wu, Z.; Puigserver, P. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. EMBO J. 2007, 26, 1913–1923. [Google Scholar] [CrossRef] [PubMed]
- Rodgers, J.T.; Lerin, C.; Haas, W.; Gygi, S.P.; Spiegelman, B.M.; Puigserver, P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 2005, 434, 113–118. [Google Scholar] [CrossRef] [PubMed]
- Yuan, Y.; Huang, S.; Wang, W.; Wang, Y.; Zhang, P.; Zhu, C.; Ding, G.; Liu, B.; Yang, T.; Zhang, A. Activation of peroxisome proliferator-activated receptor-gamma coactivator 1alpha ameliorates mitochondrial dysfunction and protects podocytes from aldosterone-induced injury. Kidney Int. 2012, 82, 771–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Funk, J.A.; Odejinmi, S.; Schnellmann, R.G. SRT1720 induces mitochondrial biogenesis and rescues mitochondrial function after oxidant injury in renal proximal tubule cells. J. Pharm. Exp. 2010, 333, 593–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chuang, P.Y.; Cai, W.; Li, X.; Fang, L.; Xu, J.; Yacoub, R.; He, J.C.; Lee, K. Reduction in podocyte SIRT1 accelerates kidney injury in aging mice. Am. J. Physiol. Ren. Physiol. 2017, 313, F621–F628. [Google Scholar] [CrossRef]
- Lan, F.; Cacicedo, J.M.; Ruderman, N.; Ido, Y. SIRT1 modulation of the acetylation status, cytosolic localization, and activity of LKB1. Possible role in AMP-activated protein kinase activation. J. Biol. Chem 2008, 283, 27628–27635. [Google Scholar] [CrossRef] [Green Version]
- Cantó, C.; Gerhart-Hines, Z.; Feige, J.N.; Lagouge, M.; Noriega, L.; Milne, J.C.; Elliott, P.J.; Puigserver, P.; Auwerx, J. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 2009, 458, 1056–1060. [Google Scholar] [CrossRef]
- Ruderman, N.B.; Xu, X.J.; Nelson, L.; Cacicedo, J.M.; Saha, A.K.; Lan, F.; Ido, Y. AMPK and SIRT1: A long-standing partnership? Am. J. Physiol. Endocrinol. Metab. 2010, 298, E751–E760. [Google Scholar] [CrossRef]
- Kume, S.; Kitada, M.; Kanasaki, K.; Maegawa, H.; Koya, D. Anti-aging molecule, Sirt1: A novel therapeutic target for diabetic nephropathy. Arch. Pharmacal Res. 2013, 36, 230–236. [Google Scholar] [CrossRef]
- Hou, S.; Zheng, F.; Li, Y.; Gao, L.; Zhang, J. The protective effect of glycyrrhizic acid on renal tubular epithelial cell injury induced by high glucose. Int. J. Mol. Sci. 2014, 15, 15026–15043. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.; Chang, B.; Zhang, Y.; Yang, P.; Liu, L. Rhein promotes the expression of SIRT1 in kidney tissues of type 2 diabetic rat. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2015, 31, 615–619. [Google Scholar] [PubMed]
- Kitada, M.; Takeda, A.; Nagai, T.; Ito, H.; Kanasaki, K.; Koya, D. Dietary restriction ameliorates diabetic nephropathy through anti-inflammatory effects and regulation of the autophagy via restoration of Sirt1 in diabetic Wistar fatty (fa/fa) rats: A model of type 2 diabetes. Exp. Diabetes Res. 2011, 2011, 908185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, D.; Cai, G.Y.; Ning, Y.C.; Wang, J.C.; Lv, Y.; Hong, Q.; Cui, S.Y.; Fu, B.; Guo, Y.N.; Chen, X.M. Alleviation of senescence and epithelial-mesenchymal transition in aging kidney by short-term caloric restriction and caloric restriction mimetics via modulation of AMPK/mTOR signaling. Oncotarget 2017, 8, 16109–16121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salih, D.A.; Brunet, A. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr. Opin. Cell Biol. 2008, 20, 126–136. [Google Scholar] [CrossRef] [Green Version]
- Lalmansingh, A.S.; Karmakar, S.; Jin, Y.; Nagaich, A.K. Multiple modes of chromatin remodeling by Forkhead box proteins. Biochim. Biophys. Acta 2012, 1819, 707–715. [Google Scholar] [CrossRef] [PubMed]
- Klotz, L.O.; Sánchez-Ramos, C.; Prieto-Arroyo, I.; Urbánek, P.; Steinbrenner, H.; Monsalve, M. Redox regulation of FoxO transcription factors. Redox Biol. 2015, 6, 51–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carlomosti, F.; D’Agostino, M.; Beji, S.; Torcinaro, A.; Rizzi, R.; Zaccagnini, G.; Maimone, B.; Di Stefano, V.; De Santa, F.; Cordisco, S.; et al. Oxidative Stress-Induced miR-200c Disrupts the Regulatory Loop Among SIRT1, FOXO1, and eNOS. Antioxid. Redox Signal. 2017, 27, 328–344. [Google Scholar] [CrossRef]
- Xu, X.; Zheng, N.; Chen, Z.; Huang, W.; Liang, T.; Kuang, H. Puerarin, isolated from Pueraria lobata (Willd.), protects against diabetic nephropathy by attenuating oxidative stress. Gene 2016, 591, 411–416. [Google Scholar] [CrossRef]
- Wu, L.; Zhang, Y.; Ma, X.; Zhang, N.; Qin, G. The effect of resveratrol on FoxO1 expression in kidneys of diabetic nephropathy rats. Mol. Biol. Rep. 2012, 39, 9085–9093. [Google Scholar] [CrossRef]
- Kume, S.; Uzu, T.; Horiike, K.; Chin-Kanasaki, M.; Isshiki, K.; Araki, S.; Sugimoto, T.; Haneda, M.; Kashiwagi, A.; Koya, D. Calorie restriction enhances cell adaptation to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged kidney. J. Clin. Investig. 2010, 120, 1043–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murtaza, G.; Khan, A.K.; Rashid, R.; Muneer, S.; Hasan, S.M.F.; Chen, J. FOXO Transcriptional Factors and Long-Term Living. Oxid. Med. Cell Longev. 2017, 2017, 3494289. [Google Scholar] [CrossRef] [PubMed]
- Brunet, A.; Sweeney, L.B.; Sturgill, J.F.; Chua, K.F.; Greer, P.L.; Lin, Y.; Tran, H.; Ross, S.E.; Mostoslavsky, R.; Cohen, H.Y.; et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004, 303, 2011–2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hariharan, N.; Maejima, Y.; Nakae, J.; Paik, J.; Depinho, R.A.; Sadoshima, J. Deacetylation of FoxO by Sirt1 Plays an Essential Role in Mediating Starvation-Induced Autophagy in Cardiac Myocytes. Circ. Res. 2010, 107, 1470–1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Meng, L.; Zhao, L.; Wang, Z.; Liu, H.; Liu, G.; Guan, G. Resveratrol ameliorates hyperglycemia-induced renal tubular oxidative stress damage via modulating the SIRT1/FOXO3a pathway. Diabetes Res. Clin. Pract. 2017, 126, 172–181. [Google Scholar] [CrossRef]
- Nakagawa, T.; Kosugi, T.; Haneda, M.; Rivard, C.J.; Long, D.A. Abnormal angiogenesis in diabetic nephropathy. Diabetes 2009, 58, 1471–1478. [Google Scholar] [CrossRef] [Green Version]
- Higgins, D.F.; Kimura, K.; Bernhardt, W.M.; Shrimanker, N.; Akai, Y.; Hohenstein, B.; Saito, Y.; Johnson, R.S.; Kretzler, M.; Cohen, C.D.; et al. Hypoxia promotes fibrogenesis in vivo via HIF-1 stimulation of epithelial-to-mesenchymal transition. J. Clin. Investig. 2007, 117, 3810–3820. [Google Scholar] [CrossRef]
- Shao, Y.; Lv, C.; Wu, C.; Zhou, Y.; Wang, Q. Mir-217 promotes inflammation and fibrosis in high glucose cultured rat glomerular mesangial cells via Sirt1/HIF-1alpha signaling pathway. Diabetes Metab. Res. Rev. 2016, 32, 534–543. [Google Scholar] [CrossRef]
- Forbes, J.M.; Coughlan, M.T.; Cooper, M.E. Oxidative stress as a major culprit in kidney disease in diabetes. Diabetes 2008, 57, 1446–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nordquist, L.; Friederich-Persson, M.; Fasching, A.; Liss, P.; Shoji, K.; Nangaku, M.; Hansell, P.; Palm, F. Activation of hypoxia-inducible factors prevents diabetic nephropathy. J. Am. Soc. Nephrol. JASN 2015, 26, 328–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gomes, P.; Fleming Outeiro, T.; Cavadas, C. Emerging Role of Sirtuin 2 in the Regulation of Mammalian Metabolism. Trends Pharmacol. Sci. 2015, 36, 756–768. [Google Scholar] [CrossRef]
- Jing, E.; Gesta, S.; Kahn, C.R. SIRT2 regulates adipocyte differentiation through FoxO1 acetylation/deacetylation. Cell Metab. 2007, 6, 105–114. [Google Scholar] [CrossRef] [Green Version]
- Krishnan, J.; Danzer, C.; Simka, T.; Ukropec, J.; Walter, K.M.; Kumpf, S.; Mirtschink, P.; Ukropcova, B.; Gasperikova, D.; Pedrazzini, T.; et al. Dietary obesity-associated Hif1α activation in adipocytes restricts fatty acid oxidation and energy expenditure via suppression of the Sirt2-NAD+ system. Genes Dev. 2012, 26, 259–270. [Google Scholar] [CrossRef] [Green Version]
- Rothgiesser, K.M.; Erener, S.; Waibel, S.; Lüscher, B.; Hottiger, M.O. SIRT2 regulates NF-κB dependent gene expression through deacetylation of p65 Lys310. J. Cell Sci. 2010, 123, 4251–4258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Mu, Y.; Zhou, X.; Ji, H.; Gao, X.; Cai, W.W.; Guan, Q.; Xu, T. SIRT2-mediated FOXO3a deacetylation drives its nuclear translocation triggering FasL-induced cell apoptosis during renal ischemia reperfusion. Apoptosis 2017, 22, 519–530. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Park, S.H.; Chang, H.C.; Shapiro, J.S.; Vassilopoulos, A.; Sawicki, K.T.; Chen, C.; Shang, M.; Burridge, P.W.; Epting, C.L.; et al. Sirtuin 2 regulates cellular iron homeostasis via deacetylation of transcription factor NRF2. J. Clin. Investig. 2017, 127, 1505–1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, W.; Hong, Y.; Chen, H.; Wu, F.; Wei, X.; Ying, W. SIRT2 mediates NADH-induced increases in Nrf2, GCL, and glutathione by modulating Akt phosphorylation in PC12 cells. FEBS Lett. 2016, 590, 2241–2255. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.P.; Zhou, L.S.; Zhao, Y.Z.; Wang, S.W.; Chen, L.L.; Liu, L.X.; Ling, Z.Q.; Hu, F.J.; Sun, Y.P.; Zhang, J.Y.; et al. Regulation of G6PD acetylation by SIRT2 and KAT9 modulates NADPH homeostasis and cell survival during oxidative stress. EMBO J. 2014, 33, 1304–1320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Y.; Li, F.; Lv, L.; Li, T.; Zhou, X.; Deng, C.X.; Guan, K.L.; Lei, Q.Y.; Xiong, Y. Oxidative stress activates SIRT2 to deacetylate and stimulate phosphoglycerate mutase. Cancer Res. 2014, 74, 3630–3642. [Google Scholar] [CrossRef] [Green Version]
- Lemos, V.; de Oliveira, R.M.; Naia, L.; Szegö, É.; Ramos, E.; Pinho, S.; Magro, F.; Cavadas, C.; Rego, A.C.; Costa, V.; et al. The NAD+-dependent deacetylase SIRT2 attenuates oxidative stress and mitochondrial dysfunction and improves insulin sensitivity in hepatocytes. Hum. Mol. Genet. 2017, 26, 4105–4117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, Y.J.; Lee, A.S.; Nguyen-Thanh, T.; Kim, D.; Kang, K.P.; Lee, S.; Park, S.K.; Kim, W. SIRT2 Regulates LPS-Induced Renal Tubular CXCL2 and CCL2 Expression. J. Am. Soc. Nephrol. JASN 2015, 26, 1549–1560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, S.-C.; Park, W.; Kang, K.; Kim, W. SIRT2 is involved in cisplatin-induced acute kidney injury through regulation of mitogen-activated protein kinase phosphatase-1. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transplant. Assoc. Eur. Ren. Assoc. 2020, 35. [Google Scholar] [CrossRef] [Green Version]
- Hebert, A.S.; Dittenhafer-Reed, K.E.; Yu, W.; Bailey, D.J.; Selen, E.S.; Boersma, M.D.; Carson, J.J.; Tonelli, M.; Balloon, A.J.; Higbee, A.J.; et al. Calorie restriction and SIRT3 trigger global reprogramming of the mitochondrial protein acetylome. Mol. Cell 2013, 49, 186–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Osborne, B.; Bentley, N.L.; Montgomery, M.K.; Turner, N. The role of mitochondrial sirtuins in health and disease. Free Radic. Biol. Med. 2016, 100, 164–174. [Google Scholar] [CrossRef]
- Perico, L.; Morigi, M.; Benigni, A. Mitochondrial Sirtuin 3 and Renal Diseases. Nephron 2016, 134, 14–19. [Google Scholar] [CrossRef]
- Bellizzi, D.; Rose, G.; Cavalcante, P.; Covello, G.; Dato, S.; De Rango, F.; Greco, V.; Maggiolini, M.; Feraco, E.; Mari, V.; et al. A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages. Genomics 2005, 85, 258–263. [Google Scholar] [CrossRef]
- Rose, G.; Dato, S.; Altomare, K.; Bellizzi, D.; Garasto, S.; Greco, V.; Passarino, G.; Feraco, E.; Mari, V.; Barbi, C.; et al. Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly. Exp. Gerontol. 2003, 38, 1065–1070. [Google Scholar] [CrossRef]
- Hirschey, M.D.; Shimazu, T.; Goetzman, E.; Jing, E.; Schwer, B.; Lombard, D.B.; Grueter, C.A.; Harris, C.; Biddinger, S.; Ilkayeva, O.R.; et al. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature 2010, 464, 121–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, M.; Li, C.M.; Ye, Z.C.; Huang, J.; Li, Y.; Lai, W.; Peng, H.; Lou, T.Q. Sirt3 modulates fatty acid oxidation and attenuates cisplatin-induced AKI in mice. J. Cell. Mol. Med. 2020, 24, 5109–5121. [Google Scholar] [CrossRef] [Green Version]
- Bell, E.L.; Emerling, B.M.; Ricoult, S.J.; Guarente, L. SirT3 suppresses hypoxia inducible factor 1α and tumor growth by inhibiting mitochondrial ROS production. Oncogene 2011, 30, 2986–2996. [Google Scholar] [CrossRef] [Green Version]
- Someya, S.; Yu, W.; Hallows, W.C.; Xu, J.; Vann, J.M.; Leeuwenburgh, C.; Tanokura, M.; Denu, J.M.; Prolla, T.A. Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell 2010, 143, 802–812. [Google Scholar] [CrossRef] [Green Version]
- Haigis, M.C.; Deng, C.X.; Finley, L.W.; Kim, H.S.; Gius, D. SIRT3 is a mitochondrial tumor suppressor: A scientific tale that connects aberrant cellular ROS, the Warburg effect, and carcinogenesis. Cancer Res. 2012, 72, 2468–2472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richter, C.; Park, J.W.; Ames, B.N. Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc. Natl. Acad. Sci. USA 1988, 85, 6465–6467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schon, E.A. Mitochondrial genetics and disease. Trends Biochem. Sci. 2000, 25, 555–560. [Google Scholar] [CrossRef]
- Cheng, Y.; Ren, X.; Gowda, A.S.; Shan, Y.; Zhang, L.; Yuan, Y.S.; Patel, R.; Wu, H.; Huber-Keener, K.; Yang, J.W.; et al. Interaction of Sirt3 with OGG1 contributes to repair of mitochondrial DNA and protects from apoptotic cell death under oxidative stress. Cell Death Dis 2013, 4, e731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lesnefsky, E.J.; Moghaddas, S.; Tandler, B.; Kerner, J.; Hoppel, C.L. Mitochondrial dysfunction in cardiac disease: Ischemia--reperfusion, aging, and heart failure. J. Mol. Cell. Cardiol. 2001, 33, 1065–1089. [Google Scholar] [CrossRef]
- Otera, H.; Mihara, K. Molecular mechanisms and physiologic functions of mitochondrial dynamics. J. Biochem. 2011, 149, 241–251. [Google Scholar] [CrossRef] [Green Version]
- Chan, D.C. Fusion and fission: Interlinked processes critical for mitochondrial health. Annu. Rev. Genet. 2012, 46, 265–287. [Google Scholar] [CrossRef] [Green Version]
- Dorn, G.W., 2nd; Song, M.; Walsh, K. Functional implications of mitofusin 2-mediated mitochondrial-SR tethering. J. Mol. Cell. Cardiol. 2015, 78, 123–128. [Google Scholar] [CrossRef] [Green Version]
- Frezza, C.; Cipolat, S.; Martins de Brito, O.; Micaroni, M.; Beznoussenko, G.V.; Rudka, T.; Bartoli, D.; Polishuck, R.S.; Danial, N.N.; De Strooper, B.; et al. OPA1 Controls Apoptotic Cristae Remodeling Independently from Mitochondrial Fusion. Cell 2006, 126, 177–189. [Google Scholar] [CrossRef] [Green Version]
- Samant, S.A.; Zhang, H.J.; Hong, Z.; Pillai, V.B.; Sundaresan, N.R.; Wolfgeher, D.; Archer, S.L.; Chan, D.C.; Gupta, M.P. SIRT3 deacetylates and activates OPA1 to regulate mitochondrial dynamics during stress. Mol. Cell Biol. 2014, 34, 807–819. [Google Scholar] [CrossRef] [Green Version]
- Morigi, M.; Perico, L.; Rota, C.; Longaretti, L.; Conti, S.; Rottoli, D.; Novelli, R.; Remuzzi, G.; Benigni, A. Sirtuin 3-dependent mitochondrial dynamic improvements protect against acute kidney injury. J. Clin. Investig. 2015, 125, 715–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qiu, X.; Brown, K.; Hirschey, M.D.; Verdin, E.; Chen, D. Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab. 2010, 12, 662–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jing, E.; Emanuelli, B.; Hirschey, M.D.; Boucher, J.; Lee, K.Y.; Lombard, D.; Verdin, E.M.; Kahn, C.R. Sirtuin-3 (Sirt3) regulates skeletal muscle metabolism and insulin signaling via altered mitochondrial oxidation and reactive oxygen species production. Proc. Natl. Acad. Sci. USA 2011, 108, 14608–14613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ogura, Y.; Kitada, M.; Monno, I.; Kanasaki, K.; Watanabe, A.; Koya, D. Renal mitochondrial oxidative stress is enhanced by the reduction of Sirt3 activity, in Zucker diabetic fatty rats. Redox Rep. Commun. Free Radic. Res. 2018, 23, 153–159. [Google Scholar] [CrossRef] [Green Version]
- Michishita, E.; Park, J.Y.; Burneskis, J.M.; Barrett, J.C.; Horikawa, I. Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins. Mol. Biol. Cell 2005, 16, 4623–4635. [Google Scholar] [CrossRef] [Green Version]
- Mathias, R.A.; Greco, T.M.; Oberstein, A.; Budayeva, H.G.; Chakrabarti, R.; Rowland, E.A.; Kang, Y.; Shenk, T.; Cristea, I.M. Sirtuin 4 is a lipoamidase regulating pyruvate dehydrogenase complex activity. Cell 2014, 159, 1615–1625. [Google Scholar] [CrossRef] [Green Version]
- Jeong, S.M.; Xiao, C.; Finley, L.W.; Lahusen, T.; Souza, A.L.; Pierce, K.; Li, Y.H.; Wang, X.; Laurent, G.; German, N.J.; et al. SIRT4 has tumor-suppressive activity and regulates the cellular metabolic response to DNA damage by inhibiting mitochondrial glutamine metabolism. Cancer Cell 2013, 23, 450–463. [Google Scholar] [CrossRef] [Green Version]
- Laurent, G.; German, N.J.; Saha, A.K.; de Boer, V.C.; Davies, M.; Koves, T.R.; Dephoure, N.; Fischer, F.; Boanca, G.; Vaitheesvaran, B.; et al. SIRT4 coordinates the balance between lipid synthesis and catabolism by repressing malonyl CoA decarboxylase. Mol. Cell 2013, 50, 686–698. [Google Scholar] [CrossRef] [Green Version]
- Lang, A.; Grether-Beck, S.; Singh, M.; Kuck, F.; Jakob, S.; Kefalas, A.; Altinoluk-Hambüchen, S.; Graffmann, N.; Schneider, M.; Lindecke, A.; et al. MicroRNA-15b regulates mitochondrial ROS production and the senescence-associated secretory phenotype through sirtuin 4/SIRT4. Aging 2016, 8, 484–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dai, Y.; Liu, S.; Li, J.; Li, J.; Lan, Y.; Nie, H.; Zuo, Y. SIRT4 suppresses the inflammatory response and oxidative stress in osteoarthritis. Am. J. Transl. Res. 2020, 12, 1965–1975. [Google Scholar] [PubMed]
- Luo, Y.X.; Tang, X.; An, X.Z.; Xie, X.M.; Chen, X.F.; Zhao, X.; Hao, D.L.; Chen, H.Z.; Liu, D.P. SIRT4 accelerates Ang II-induced pathological cardiac hypertrophy by inhibiting manganese superoxide dismutase activity. Eur. Heart J. 2017, 38, 1389–1398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tarantino, G.; Finelli, C.; Scopacasa, F.; Pasanisi, F.; Contaldo, F.; Capone, D.; Savastano, S. Circulating levels of sirtuin 4, a potential marker of oxidative metabolism, related to coronary artery disease in obese patients suffering from NAFLD, with normal or slightly increased liver enzymes. Oxid. Med. Cell Longev. 2014, 2014, 920676. [Google Scholar] [CrossRef] [Green Version]
- Shi, J.X.; Wang, Q.J.; Li, H.; Huang, Q. SIRT4 overexpression protects against diabetic nephropathy by inhibiting podocyte apoptosis. Exp. Ther. Med. 2017, 13, 342–348. [Google Scholar] [CrossRef] [Green Version]
- Kumar, S.; Lombard, D.B. Functions of the sirtuin deacylase SIRT5 in normal physiology and pathobiology. Crit. Rev. Biochem. Mol. Biol. 2018, 53, 311–334. [Google Scholar] [CrossRef]
- Bringman-Rodenbarger, L.R.; Guo, A.H.; Lyssiotis, C.A.; Lombard, D.B. Emerging Roles for SIRT5 in Metabolism and Cancer. Antioxid. Redox Signal. 2018, 28, 677–690. [Google Scholar] [CrossRef]
- Rardin, M.J.; He, W.; Nishida, Y.; Newman, J.C.; Carrico, C.; Danielson, S.R.; Guo, A.; Gut, P.; Sahu, A.K.; Li, B.; et al. SIRT5 regulates the mitochondrial lysine succinylome and metabolic networks. Cell Metab. 2013, 18, 920–933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, J.; Chen, Y.; Tishkoff, D.X.; Peng, C.; Tan, M.; Dai, L.; Xie, Z.; Zhang, Y.; Zwaans, B.M.; Skinner, M.E.; et al. SIRT5-mediated lysine desuccinylation impacts diverse metabolic pathways. Mol. Cell 2013, 50, 919–930. [Google Scholar] [CrossRef] [Green Version]
- Nakagawa, T.; Lomb, D.J.; Haigis, M.C.; Guarente, L. SIRT5 Deacetylates carbamoyl phosphate synthetase 1 and regulates the urea cycle. Cell 2009, 137, 560–570. [Google Scholar] [CrossRef] [Green Version]
- Ogura, M.; Nakamura, Y.; Tanaka, D.; Zhuang, X.; Fujita, Y.; Obara, A.; Hamasaki, A.; Hosokawa, M.; Inagaki, N. Overexpression of SIRT5 confirms its involvement in deacetylation and activation of carbamoyl phosphate synthetase 1. Biochem. Biophys. Res. Commun. 2010, 393, 73–78. [Google Scholar] [CrossRef] [PubMed]
- Bobermin, L.D.; Wartchow, K.M.; Flores, M.P.; Leite, M.C.; Quincozes-Santos, A.; Gonçalves, C.A. Ammonia-induced oxidative damage in neurons is prevented by resveratrol and lipoic acid with participation of heme oxygenase 1. Neurotoxicology 2015, 49, 28–35. [Google Scholar] [CrossRef] [PubMed]
- Lin, Z.F.; Xu, H.B.; Wang, J.Y.; Lin, Q.; Ruan, Z.; Liu, F.B.; Jin, W.; Huang, H.H.; Chen, X. SIRT5 desuccinylates and activates SOD1 to eliminate ROS. Biochem. Biophys. Res. Commun. 2013, 441, 191–195. [Google Scholar] [CrossRef] [PubMed]
- Schlicker, C.; Gertz, M.; Papatheodorou, P.; Kachholz, B.; Becker, C.F.; Steegborn, C. Substrates and regulation mechanisms for the human mitochondrial sirtuins Sirt3 and Sirt5. J. Mol. Biol. 2008, 382, 790–801. [Google Scholar] [CrossRef] [PubMed]
- Liang, F.; Wang, X.; Ow, S.H.; Chen, W.; Ong, W.C. Sirtuin 5 is Anti-apoptotic and Anti-oxidative in Cultured SH-EP Neuroblastoma Cells. Neurotox. Res. 2017, 31, 63–76. [Google Scholar] [CrossRef]
- Guedouari, H.; Daigle, T.; Scorrano, L.; Hebert-Chatelain, E. Sirtuin 5 protects mitochondria from fragmentation and degradation during starvation. Biochim. Biophys. Acta. Mol. Cell Res. 2017, 1864, 169–176. [Google Scholar] [CrossRef]
- Li, W.; Yang, Y.; Li, Y.; Zhao, Y.; Jiang, H. Sirt5 Attenuates Cisplatin-Induced Acute Kidney Injury through Regulation of Nrf2/HO-1 and Bcl-2. BioMed Res. Int. 2019, 2019, 4745132. [Google Scholar] [CrossRef]
- Chiba, T.; Peasley, K.D.; Cargill, K.R.; Maringer, K.V.; Bharathi, S.S.; Mukherjee, E.; Zhang, Y.; Holtz, A.; Basisty, N.; Yagobian, S.D.; et al. Sirtuin 5 Regulates Proximal Tubule Fatty Acid Oxidation to Protect against AKI. J. Am. Soc. Nephrol. JASN 2019, 30, 2384–2398. [Google Scholar] [CrossRef]
- Haschler, T.N. Mitochondrial Dysfunction and the Role of Sirt5 in Renal Disease; UCL (University College London): London, UK, 2020. [Google Scholar]
- Tasselli, L.; Zheng, W.; Chua, K.F. SIRT6: Novel Mechanisms and Links to Aging and Disease. Trends Endocrinol. Metab. TEM 2017, 28, 168–185. [Google Scholar] [CrossRef] [Green Version]
- Kanfi, Y.; Naiman, S.; Amir, G.; Peshti, V.; Zinman, G.; Nahum, L.; Bar-Joseph, Z.; Cohen, H.Y. The sirtuin SIRT6 regulates lifespan in male mice. Nature 2012, 483, 218–221. [Google Scholar] [CrossRef]
- Mostoslavsky, R.; Chua, K.F.; Lombard, D.B.; Pang, W.W.; Fischer, M.R.; Gellon, L.; Liu, P.; Mostoslavsky, G.; Franco, S.; Murphy, M.M.; et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 2006, 124, 315–329. [Google Scholar] [CrossRef] [Green Version]
- Dominy, J.E., Jr.; Lee, Y.; Jedrychowski, M.P.; Chim, H.; Jurczak, M.J.; Camporez, J.P.; Ruan, H.B.; Feldman, J.; Pierce, K.; Mostoslavsky, R.; et al. The deacetylase Sirt6 activates the acetyltransferase GCN5 and suppresses hepatic gluconeogenesis. Mol. Cell 2012, 48, 900–913. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Hou, T.; Gao, T.; Lu, X.; Yang, Q.; Zhu, Q.; Li, Z.; Liu, C.; Mu, G.; Liu, G.; et al. p53 cooperates with SIRT6 to regulate cardiolipin de novo biosynthesis. Cell Death Dis 2018, 9, 941. [Google Scholar] [CrossRef] [PubMed]
- Hu, S.; Liu, H.; Ha, Y.; Luo, X.; Motamedi, M.; Gupta, M.P.; Ma, J.X.; Tilton, R.G.; Zhang, W. Posttranslational modification of Sirt6 activity by peroxynitrite. Free Radic. Biol. Med. 2015, 79, 176–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balestrieri, M.L.; Rizzo, M.R.; Barbieri, M.; Paolisso, P.; D’Onofrio, N.; Giovane, A.; Siniscalchi, M.; Minicucci, F.; Sardu, C.; D’Andrea, D.; et al. Sirtuin 6 expression and inflammatory activity in diabetic atherosclerotic plaques: Effects of incretin treatment. Diabetes 2015, 64, 1395–1406. [Google Scholar] [CrossRef] [Green Version]
- Mao, Z.; Hine, C.; Tian, X.; Van Meter, M.; Au, M.; Vaidya, A.; Seluanov, A.; Gorbunova, V. SIRT6 promotes DNA repair under stress by activating PARP1. Science 2011, 332, 1443–1446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cardus, A.; Uryga, A.K.; Walters, G.; Erusalimsky, J.D. SIRT6 protects human endothelial cells from DNA damage, telomere dysfunction, and senescence. Cardiovasc. Res. 2013, 97, 571–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.X.; Wang, X.L.; Tong, M.M.; Gan, L.; Chen, H.; Wu, S.S.; Chen, J.X.; Li, R.L.; Wu, Y.; Zhang, H.Y.; et al. SIRT6 protects cardiomyocytes against ischemia/reperfusion injury by augmenting FoxO3α-dependent antioxidant defense mechanisms. Basic Res. Cardiol. 2016, 111, 13. [Google Scholar] [CrossRef]
- Pan, H.; Guan, D.; Liu, X.; Li, J.; Wang, L.; Wu, J.; Zhou, J.; Zhang, W.; Ren, R.; Zhang, W.; et al. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res. 2016, 26, 190–205. [Google Scholar] [CrossRef]
- Huang, W.; Liu, H.; Zhu, S.; Woodson, M.; Liu, R.; Tilton, R.G.; Miller, J.D.; Zhang, W. Sirt6 deficiency results in progression of glomerular injury in the kidney. Aging 2017, 9, 1069–1083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Wang, L.; Meng, L.; Cao, G.; Wu, Y. Sirtuin 6 overexpression relieves sepsis-induced acute kidney injury by promoting autophagy. Cell Cycle 2019, 18, 425–436. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Xu, K.; Zhang, N.; Amador, G.; Wang, Y.; Zhao, S.; Li, L.; Qiu, Y.; Wang, Z. Overexpressed SIRT6 attenuates cisplatin-induced acute kidney injury by inhibiting ERK1/2 signaling. Kidney Int. 2018, 93, 881–892. [Google Scholar] [CrossRef]
- Grabowska, W.; Sikora, E.; Bielak-Zmijewska, A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology 2017, 18, 447–476. [Google Scholar] [CrossRef] [Green Version]
- Barber, M.F.; Michishita-Kioi, E.; Xi, Y.; Tasselli, L.; Kioi, M.; Moqtaderi, Z.; Tennen, R.I.; Paredes, S.; Young, N.L.; Chen, K.; et al. SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation. Nature 2012, 487, 114–118. [Google Scholar] [CrossRef]
- Kiran, S.; Anwar, T.; Kiran, M.; Ramakrishna, G. Sirtuin 7 in cell proliferation, stress and disease: Rise of the Seventh Sirtuin! Cell. Signal. 2015, 27, 673–682. [Google Scholar] [CrossRef] [PubMed]
- Ryu, D.; Jo, Y.S.; Lo Sasso, G.; Stein, S.; Zhang, H.; Perino, A.; Lee, J.U.; Zeviani, M.; Romand, R.; Hottiger, M.O.; et al. A SIRT7-dependent acetylation switch of GABPβ1 controls mitochondrial function. Cell Metab. 2014, 20, 856–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, W.; Fu, Y.C.; Zhou, X.H.; Chen, C.J.; Wang, X.; Lin, R.B.; Wang, W. Effects of resveratrol on H(2)O(2)-induced apoptosis and expression of SIRTs in H9c2 cells. J. Cell Biochem. 2009, 107, 741–747. [Google Scholar] [CrossRef] [PubMed]
- Vakhrusheva, O.; Smolka, C.; Gajawada, P.; Kostin, S.; Boettger, T.; Kubin, T.; Braun, T.; Bober, E. Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice. Circ. Res. 2008, 102, 703–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, B.L. SIRT7 and hepatic lipid metabolism. Front. Cell Dev. Biol. 2015, 3, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miyasato, Y.; Yoshizawa, T.; Sato, Y.; Nakagawa, T.; Miyasato, Y.; Kakizoe, Y.; Kuwabara, T.; Adachi, M.; Ianni, A.; Braun, T.; et al. Sirtuin 7 Deficiency Ameliorates Cisplatin-induced Acute Kidney Injury Through Regulation of the Inflammatory Response. Sci. Rep. 2018, 8, 5927. [Google Scholar] [CrossRef]
- Hubbi, M.E.; Hu, H.; Kshitiz; Gilkes, D.M.; Semenza, G.L. Sirtuin-7 inhibits the activity of hypoxia-inducible factors. J. Biol. Chem. 2013, 288, 20768–20775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, T.; Chi, Y.; Ren, Y.; Du, C.; Shi, Y.; Li, Y. Resveratrol Reduces Oxidative Stress and Apoptosis in Podocytes via Sir2-Related Enzymes, Sirtuins1 (SIRT1)/Peroxisome Proliferator-Activated Receptor gamma Co-Activator 1alpha (PGC-1alpha) Axis. Med. Sci. Monit 2019, 25, 1220–1231. [Google Scholar] [CrossRef] [PubMed]
- Kitada, M.; Koya, D. Renal protective effects of resveratrol. Oxid. Med. Cell Longev. 2013, 2013, 568093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, D.F.; You, N.; Wu, X.M.; Xu, J.R.; Hu, A.P.; Ye, X.L.; Zhu, Q.; Jiang, X.Q.; Miao, H.; Liu, C.; et al. Resveratrol attenuates renal hypertrophy in early-stage diabetes by activating AMPK. Am. J. Nephrol. 2010, 31, 363–374. [Google Scholar] [CrossRef]
- Gan, Y.; Tao, S.; Cao, D.; Xie, H.; Zeng, Q. Protection of resveratrol on acute kidney injury in septic rats. Hum. Exp. Toxicol. 2017, 36, 1015–1022. [Google Scholar] [CrossRef]
- Pan, W.; Yu, H.; Huang, S.; Zhu, P. Resveratrol Protects against TNF-alpha-Induced Injury in Human Umbilical Endothelial Cells through Promoting Sirtuin-1-Induced Repression of NF-KB and p38 MAPK. PLoS ONE 2016, 11, e0147034. [Google Scholar] [CrossRef] [Green Version]
- Xia, N.; Strand, S.; Schlufter, F.; Siuda, D.; Reifenberg, G.; Kleinert, H.; Forstermann, U.; Li, H. Role of SIRT1 and FOXO factors in eNOS transcriptional activation by resveratrol. Nitric Oxide 2013, 32, 29–35. [Google Scholar] [CrossRef]
- Wang, X.; Gao, Y.; Tian, N.; Zou, D.; Shi, Y.; Zhang, N. Astragaloside IV improves renal function and fibrosis via inhibition of miR-21-induced podocyte dedifferentiation and mesangial cell activation in diabetic mice. Drug Des. Devel. 2018, 12, 2431–2442. [Google Scholar] [CrossRef] [Green Version]
- Mitchell, S.J.; Martin-Montalvo, A.; Mercken, E.M.; Palacios, H.H.; Ward, T.M.; Abulwerdi, G.; Minor, R.K.; Vlasuk, G.P.; Ellis, J.L.; Sinclair, D.A.; et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep. 2014, 6, 836–843. [Google Scholar] [CrossRef] [Green Version]
- Mercken, E.M.; Mitchell, S.J.; Martin-Montalvo, A.; Minor, R.K.; Almeida, M.; Gomes, A.P.; Scheibye-Knudsen, M.; Palacios, H.H.; Licata, J.J.; Zhang, Y.; et al. SRT2104 extends survival of male mice on a standard diet and preserves bone and muscle mass. Aging Cell 2014, 13, 787–796. [Google Scholar] [CrossRef]
- Roberts, A.B.; Tian, F.; Byfield, S.D.; Stuelten, C.; Ooshima, A.; Saika, S.; Flanders, K.C. Smad3 is key to TGF-beta-mediated epithelial-to-mesenchymal transition, fibrosis, tumor suppression and metastasis. Cytokine Growth Factor Rev. 2006, 17, 19–27. [Google Scholar] [CrossRef]
- Nagahisa, T.; Saisho, Y. Cardiorenal Protection: Potential of SGLT2 Inhibitors and GLP-1 Receptor Agonists in the Treatment of Type 2 Diabetes. Diabetes Ther. Res. Treat. Educ. Diabetes Relat. Disord. 2019, 10, 1733–1752. [Google Scholar] [CrossRef]
- Packer, M. Role of ketogenic starvation sensors in mediating the renal protective effects of SGLT2 inhibitors in type 2 diabetes. J. Diabetes Its Complicat. 2020, 107647. [Google Scholar] [CrossRef] [PubMed]
- Ogura, Y.; Kitada, M.; Xu, J.; Monno, I.; Koya, D. CD38 inhibition by apigenin ameliorates mitochondrial oxidative stress through restoration of the intracellular NAD(+)/NADH ratio and Sirt3 activity in renal tubular cells in diabetic rats. Aging 2020, 12, 11325–11336. [Google Scholar] [CrossRef] [PubMed]
- Cucuz, V.; Cvejic, J.; Bukarica, L. Clinical trials of resveratrol efficacy and safety. Vojnosanit. Pregl. 2021, 6. [Google Scholar] [CrossRef]
- Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; García-Almagro, F.J.; Avilés-Plaza, F.; Parra, S.; Yáñez-Gascón, M.J.; Ruiz-Ros, J.A.; García-Conesa, M.T.; Tomás-Barberán, F.A.; et al. Consumption of a grape extract supplement containing resveratrol decreases oxidized LDL and ApoB in patients undergoing primary prevention of cardiovascular disease: A triple-blind, 6-month follow-up, placebo-controlled, randomized trial. Mol. Nutr. Food Res. 2012, 56, 810–821. [Google Scholar] [CrossRef]
- Simental-Mendía, L.E.; Guerrero-Romero, F. Effect of resveratrol supplementation on lipid profile in subjects with dyslipidemia: A randomized double-blind, placebo-controlled trial. Nutrition 2019, 58, 7–10. [Google Scholar] [CrossRef]
- Fogacci, F.; Tocci, G.; Presta, V.; Fratter, A.; Borghi, C.; Cicero, A.F.G. Effect of resveratrol on blood pressure: A systematic review and meta-analysis of randomized, controlled, clinical trials. Crit. Rev. Food Sci. Nutr. 2019, 59, 1605–1618. [Google Scholar] [CrossRef]
- Kitada, M.; Ogura, Y.; Monno, I.; Koya, D.J.N. Supplementation with Red Wine Extract Increases Insulin Sensitivity and Peripheral Blood Mononuclear Sirt1 Expression in Nondiabetic Humans. Nutrients 2020, 12, 3108. [Google Scholar] [CrossRef] [PubMed]
- Kitada, M.; Ogura, Y.; Maruki-Uchida, H.; Sai, M.; Suzuki, T.; Kanasaki, K.; Hara, Y.; Seto, H.; Kuroshima, Y.; Monno, I.; et al. The Effect of Piceatannol from Passion Fruit (Passiflora edulis) Seeds on Metabolic Health in Humans. Nutrients 2017, 9, 1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoshino, M.; Yoshino, J.; Kayser, B.D.; Patti, G.J.; Franczyk, M.P.; Mills, K.F.; Sindelar, M.; Pietka, T.; Patterson, B.W.; Imai, S.-I.; et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science 2021, 372, 1224. [Google Scholar] [CrossRef] [PubMed]
- Yasuda, I.; Hasegawa, K.; Sakamaki, Y.; Muraoka, H.; Kawaguchi, T.; Kusahana, E.; Ono, T.; Kanda, T.; Tokuyama, H.; Wakino, S.; et al. Pre-emptive Short-term Nicotinamide Mononucleotide Treatment in a Mouse Model of Diabetic Nephropathy. J. Am. Soc. Nephrol. 2021, 32, 1355–1370. [Google Scholar] [CrossRef] [PubMed]
Localization | Catalytic Activity | |
---|---|---|
Sirt1 | Nucleus and Cytoplasm | Deacetylation |
Sirt2 | Cytoplasm and Nucleus | Deacetylation |
Sirt3 | Mitochondria | Deacetylation |
Sirt4 | Mitochondria | ADP-ribosylation Deacetylase |
Sirt5 | Mitochondria | Desuccinylation Demalonylation Deglutarylation Deacetylation |
Sirt6 | Nucleus and Cytoplasm | Deacetylation ADP-ribosylation |
Sirt7 | Nucleus and Cytoplasm | Deacetylation |
Target Molecules | Kidney Model | Activetor | |
---|---|---|---|
Sirt1 | NF-ĸB SOD2 CAT Nrf2 p53 eNOS p66Shc PGC-1α LKB1/AMPK FoxO1 FoxO3a HIF-1α | Diabetic kidney Aging kidney HG cultured tubular epithelial cell HG cultured podocyte Human embryonic kidney cells | CR Resveratrol VAS-IV Glycyrrhizic acid Puerarin SRT1720 SRT2104 SRT3025 SGLT2 inhibitors |
Sirt2 | NF-ĸB SOD2 PGC-1α HIF-1α Nrf2 G6PD PGAM2 ERK1 DRP1 Mfn2 TFAM MKP-1 | Renal Ischemia/Reperfusion model LPS nephropathy Cisplatin-induced Acute kidney injury | CR |
Target Molecules | Kidney Model | Activator | |
---|---|---|---|
Sirt3 | OGG1 OPA1 SOD2 IDH2 CAT AMPK HIF-1α | Cisplatin-induced Acute kidney injury Renal Ischemia/Reperfusion model Sepsis-induced Acute kidney injury Diabetic kidney | Honokiol Apigenin AICAR ALCAR |
Sirt4 | GDH PDC malonylCoA decarboxylase SOD CAT | Diabetic kidney Aging kidney HG cultured podocyte | CR |
Sirt5 | Mitochondrial Complex 1 Mitochondrial Complex 2 ATP synthase CPS1 SOD1 Cytochrome C Nrf2 | Cisplatin-induced Acute kidney injury Renal Ischemia/Reperfusion model Folate nephritis | Fasting |
Target Molecules | Kidney Model | Activator | |
---|---|---|---|
Sirt6 | NF-ĸB PGC-1α p53 FoxO1 FoxO3a AMPK eNOS Nrf2 | LPS nephropathy Cisplatin-induced Acute kidney injury | CR |
Sirt7 | GABPb1 NF-ĸB HIF-1α | Cisplatin-induced Acute kidney injury | CR Resveratrol |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ogura, Y.; Kitada, M.; Koya, D. Sirtuins and Renal Oxidative Stress. Antioxidants 2021, 10, 1198. https://doi.org/10.3390/antiox10081198
Ogura Y, Kitada M, Koya D. Sirtuins and Renal Oxidative Stress. Antioxidants. 2021; 10(8):1198. https://doi.org/10.3390/antiox10081198
Chicago/Turabian StyleOgura, Yoshio, Munehiro Kitada, and Daisuke Koya. 2021. "Sirtuins and Renal Oxidative Stress" Antioxidants 10, no. 8: 1198. https://doi.org/10.3390/antiox10081198
APA StyleOgura, Y., Kitada, M., & Koya, D. (2021). Sirtuins and Renal Oxidative Stress. Antioxidants, 10(8), 1198. https://doi.org/10.3390/antiox10081198