p66Shc in Cardiovascular Pathology
Abstract
:1. Introduction
2. Reactive Oxygen Species, Aging, and Oxidative Stress
3. Reactive Oxygen Species—Neutralization, Necessity, and Pathology
4. p66Shc Discovery—Aging Research
5. The ShcA Family
6. p66Shc Expression and Localization
7. p66Shc Signaling Overview
8. Classical p66Shc Mitochondrial ROS Activity Pathway
8.1. Absent of Cell Stress, Peroxiredoxin 1 Prevents p66Shc Mitochondrial Translocation via Cytoplasmic Sequestration
8.2. Cell Stress Causes Peroxiredoxin 1-p66Shc Dissociation, Allowing Protein Kinase c Family Members to Transduce Cell Stress Signals by Phosphorylating p66Shc at Ser36
8.3. Prolyl Isomerase 1 Interacts with p66Shc, Causing Conformational Changes That Prime p66Shc for Mitochondrial Translocation
8.4. p66Shc Is Dephosphorylated by Protein Phosphatase 2A to Allow Mitochondrial Translocation
8.5. p66Shc Enters the Intermembrane Space via Interactions with Mitochondrial Translocase of the Outer Membrane
8.6. p66Shc Interacts with Cytochrome c, Causing H2O2 Accumulation, Permeability Transition Pore Formation, and Apoptosis
9. Recent Advances in p66Shc Mitochondrial Function
9.1. p66Shc Oligomerization Status and Structure
9.2. p66Shc ROS Identity and Regulation
9.3. p66Shc Superoxide Anion Mechanism
9.4. p66Shc-Cytochrome c Interactions
9.5. Revised p66Shc Mitochondrial Function Model
10. p66Shc’s Mitochondrial ROS Activity Role in Cardiovascular Pathology
10.1. p66Shc in Ischemia/Reperfusion Injuries
10.2. p66Shc in Endothelial Dysfunction
11. Future Perspectives and Summary
Author Contributions
Funding
Conflicts of Interest
References
- Zheng, Y.; Zhang, C.; Croucher, D.R.; Soliman, M.A.; St-Denis, N.; Pasculescu, A.; Taylor, L.; Tate, S.A.; Hardy, W.R.; Colwill, K.; et al. Temporal regulation of EGF signalling networks by the scaffold protein Shc1. Nature 2013, 499, 166–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Sweet, D.T.; Irani-Tehrani, M.; Maeda, N.; Tzima, E. Shc coordinates signals from intercellular junctions and integrins to regulate flow-induced inflammation. J. Cell Biol. 2008, 182, 185–196. [Google Scholar] [CrossRef]
- Soliman, M.A.; Abdel Rahman, A.M.; Lamming, D.W.; Birsoy, K.; Pawling, J.; Frigolet, M.E.; Lu, H.; Fantus, I.G.; Pasculescu, A.; Zheng, Y.; et al. The adaptor protein p66Shc inhibits mTOR-dependent anabolic metabolism. Sci. Signal 2014, 7, ra17. [Google Scholar] [CrossRef] [Green Version]
- Migliaccio, E.; Giorgio, M.; Mele, S.; Pelicci, G.; Reboldi, P.; Pandolfi, P.P.; Lanfrancone, L.; Pelicci, P.G. The p66shc adaptor protein controls oxidative stress response and life span in mammals. Nature 1999, 402, 309–313. [Google Scholar] [CrossRef] [PubMed]
- Giorgio, M.; Migliaccio, E.; Orsini, F.; Paolucci, D.; Moroni, M.; Contursi, C.; Pelliccia, G.; Luzi, L.; Minucci, S.; Marcaccio, M.; et al. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell 2005, 122, 221–233. [Google Scholar] [CrossRef]
- Di Lisa, F.; Kaludercic, N.; Carpi, A.; Menabo, R.; Giorgio, M. Mitochondrial pathways for ROS formation and myocardial injury: The relevance of p66(Shc) and monoamine oxidase. Basic Res. Cardiol. 2009, 104, 131–139. [Google Scholar] [CrossRef]
- Galimov, E.R. The Role of p66shc in Oxidative Stress and Apoptosis. Acta Nat. 2010, 2, 44–51. [Google Scholar] [CrossRef]
- Cano Sanchez, M.; Lancel, S.; Boulanger, E.; Neviere, R. Targeting Oxidative Stress and Mitochondrial Dysfunction in the Treatment of Impaired Wound Healing: A Systematic Review. Antioxidants 2018, 7, 98. [Google Scholar] [CrossRef] [Green Version]
- D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef] [PubMed]
- Silvia, B. Preconditioning and Shear Stress in the Microcirculation in Ischemia-Reperfusion Injury. Curr. Cardiol. Rev. 2006, 2, 237–245. [Google Scholar] [CrossRef]
- Franzeck, F.C.; Hof, D.; Spescha, R.D.; Hasun, M.; Akhmedov, A.; Steffel, J.; Shi, Y.; Cosentino, F.; Tanner, F.C.; von Eckardstein, A.; et al. Expression of the aging gene p66Shc is increased in peripheral blood monocytes of patients with acute coronary syndrome but not with stable coronary artery disease. Atherosclerosis 2012, 220, 282–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noda, Y.; Yamagishi, S.; Matsui, T.; Ueda, S.; Ueda, S.; Jinnouchi, Y.; Hirai, Y.; Imaizumi, T. The p66shc gene expression in peripheral blood monocytes is increased in patients with coronary artery disease. Clin. Cardiol. 2010, 33, 548–552. [Google Scholar] [CrossRef] [PubMed]
- Spescha, R.D.; Klohs, J.; Semerano, A.; Giacalone, G.; Derungs, R.S.; Reiner, M.F.; Rodriguez Gutierrez, D.; Mendez-Carmona, N.; Glanzmann, M.; Savarese, G.; et al. Post-ischaemic silencing of p66Shc reduces ischaemia/reperfusion brain injury and its expression correlates to clinical outcome in stroke. Eur. Heart J. 2015, 36, 1590–1600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bugger, H.; Pfeil, K. Mitochondrial ROS in myocardial ischemia reperfusion and remodeling. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165768. [Google Scholar] [CrossRef]
- Shaw, P.X.; Werstuck, G.; Chen, Y. Oxidative stress and aging diseases. Oxid. Med. Cell. Longev. 2014, 2014, 569146. [Google Scholar] [CrossRef] [PubMed]
- Ghezzi, P.; Jaquet, V.; Marcucci, F.; Schmidt, H. The oxidative stress theory of disease: Levels of evidence and epistemological aspects. Br. J. Pharm. 2017, 174, 1784–1796. [Google Scholar] [CrossRef]
- Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956, 11, 298–300. [Google Scholar] [CrossRef] [Green Version]
- Shields, H.J.; Traa, A.; Van Raamsdonk, J.M. Beneficial and Detrimental Effects of Reactive Oxygen Species on Lifespan: A Comprehensive Review of Comparative and Experimental Studies. Front. Cell Dev. Biol. 2021, 9, 628157. [Google Scholar] [CrossRef]
- Milkovic, L.; Cipak Gasparovic, A.; Cindric, M.; Mouthuy, P.A.; Zarkovic, N. Short Overview of ROS as Cell Function Regulators and Their Implications in Therapy Concepts. Cells 2019, 8, 793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santos, A.L.; Sinha, S.; Lindner, A.B. The Good, the Bad, and the Ugly of ROS: New Insights on Aging and Aging-Related Diseases from Eukaryotic and Prokaryotic Model Organisms. Oxid. Med. Cell Longev. 2018, 2018, 1941285. [Google Scholar] [CrossRef] [PubMed]
- Pham-Huy, L.A.; He, H.; Pham-Huy, C. Free radicals, antioxidants in disease and health. Int. J. Biomed. Sci. 2008, 4, 89–96. [Google Scholar]
- 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] [PubMed] [Green Version]
- Winterbourn, C.C. Reconciling the chemistry and biology of reactive oxygen species. Nat. Chem. Biol. 2008, 4, 278–286. [Google Scholar] [CrossRef] [PubMed]
- Mittler, R. ROS Are Good. Trends Plant Sci. 2017, 22, 11–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Marzo, N.; Chisci, E.; Giovannoni, R. The Role of Hydrogen Peroxide in Redox-Dependent Signaling: Homeostatic and Pathological Responses in Mammalian Cells. Cells 2018, 7, 156. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- Sutton, H.C.; Winterbourn, C.C. On the participation of higher oxidation states of iron and copper in Fenton reactions. Free Radic Biol Med. 1989, 6, 53–60. [Google Scholar] [CrossRef]
- Mylonas, C.; Kouretas, D. Lipid peroxidation and tissue damage. Vivo 1999, 13, 295–309. [Google Scholar]
- Sohal, R.S. Role of oxidative stress and protein oxidation in the aging process. Free Radic. Biol. Med. 2002, 33, 37–44. [Google Scholar] [CrossRef]
- Kregel, K.C.; Zhang, H.J. An integrated view of oxidative stress in aging: Basic mechanisms, functional effects, and pathological considerations. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 292, R18–R36. [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]
- Syslova, K.; Bohmova, A.; Mikoska, M.; Kuzma, M.; Pelclova, D.; Kacer, P. Multimarker screening of oxidative stress in aging. Oxid. Med. Cell. Longev. 2014, 2014, 562860. [Google Scholar] [CrossRef] [Green Version]
- Park, C.B.; Larsson, N.G. Mitochondrial DNA mutations in disease and aging. J. Cell Biol. 2011, 193, 809–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jha, R.; Rizvi, S.I. Carbonyl formation in erythrocyte membrane proteins during aging in humans. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech. Repub. 2011, 155, 39–42. [Google Scholar] [CrossRef] [Green Version]
- Moskovitz, J.; Oien, D.B. Protein carbonyl and the methionine sulfoxide reductase system. Antioxid. Redox Signal. 2010, 12, 405–415. [Google Scholar] [CrossRef]
- Moskalev, A.A.; Shaposhnikov, M.V.; Plyusnina, E.N.; Zhavoronkov, A.; Budovsky, A.; Yanai, H.; Fraifeld, V.E. The role of DNA damage and repair in aging through the prism of Koch-like criteria. Ageing Res. Rev. 2013, 12, 661–684. [Google Scholar] [CrossRef] [PubMed]
- Massudi, H.; Grant, R.; Braidy, N.; Guest, J.; Farnsworth, B.; Guillemin, G.J. Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PLoS ONE 2012, 7, e42357. [Google Scholar] [CrossRef]
- Nguyen, G.T.; Green, E.R.; Mecsas, J. Neutrophils to the ROScue: Mechanisms of NADPH Oxidase Activation and Bacterial Resistance. Front. Cell Infect. Microbiol. 2017, 7, 373. [Google Scholar] [CrossRef]
- Beckman, K.B.; Ames, B.N. The free radical theory of aging matures. Physiol. Rev. 1998, 78, 547–581. [Google Scholar] [CrossRef] [Green Version]
- Lambeth, J.D. NOX enzymes and the biology of reactive oxygen. Nat. Rev. Immunol. 2004, 4, 181–189. [Google Scholar] [CrossRef]
- Shih, J.C.; Chen, K.; Ridd, M.J. Monoamine oxidase: From genes to behavior. Annu. Rev. Neurosci. 1999, 22, 197–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Battelli, M.G.; Polito, L.; Bortolotti, M.; Bolognesi, A. Xanthine Oxidoreductase in Drug Metabolism: Beyond a Role as a Detoxifying Enzyme. Curr. Med. Chem. 2016, 23, 4027–4036. [Google Scholar] [CrossRef] [Green Version]
- McDonnell, A.M.; Dang, C.H. Basic review of the cytochrome p450 system. J. Adv. Pract. Oncol. 2013, 4, 263–268. [Google Scholar] [CrossRef] [PubMed]
- Lismont, C.; Nordgren, M.; Van Veldhoven, P.P.; Fransen, M. Redox interplay between mitochondria and peroxisomes. Front. Cell Dev. Biol. 2015, 3, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Meo, S.; Reed, T.T.; Venditti, P.; Victor, V.M. Role of ROS and RNS Sources in Physiological and Pathological Conditions. Oxid. Med. Cell. Longev. 2016, 2016, 1245049. [Google Scholar] [CrossRef] [PubMed]
- Van Raamsdonk, J.M.; Hekimi, S. Reactive Oxygen Species and Aging in Caenorhabditis elegans: Causal or Casual Relationship? Antioxid. Redox Signal. 2010, 13, 1911–1953. [Google Scholar] [CrossRef]
- Balaban, R.S.; Nemoto, S.; Finkel, T. Mitochondria, oxidants, and aging. Cell 2005, 120, 483–495. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Vazquez, E.J.; Moghaddas, S.; Hoppel, C.L.; Lesnefsky, E.J. Production of reactive oxygen species by mitochondria: Central role of complex III. J. Biol. Chem. 2003, 278, 36027–36031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kushnareva, Y.; Murphy, A.N.; Andreyev, A. Complex I-mediated reactive oxygen species generation: Modulation by cytochrome c and NAD(P)+ oxidation-reduction state. Biochem. J. 2002, 368, 545–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Forman, H.J.; Ursini, F.; Maiorino, M. An overview of mechanisms of redox signaling. J. Mol. Cell. Cardiol. 2014, 73, 2–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Korshunov, S.S.; Krasnikov, B.F.; Pereverzev, M.O.; Skulachev, V.P. The antioxidant functions of cytochrome c. FEBS Lett. 1999, 462, 192–198. [Google Scholar] [CrossRef] [Green Version]
- Sedlak, E.; Fabian, M.; Robinson, N.C.; Musatov, A. Ferricytochrome c protects mitochondrial cytochrome c oxidase against hydrogen peroxide-induced oxidative damage. Free Radic. Biol. Med. 2010, 49, 1574–1581. [Google Scholar] [CrossRef] [Green Version]
- Pasdois, P.; Parker, J.E.; Griffiths, E.J.; Halestrap, A.P. The role of oxidized cytochrome c in regulating mitochondrial reactive oxygen species production and its perturbation in ischaemia. Biochem. J. 2011, 436, 493–505. [Google Scholar] [CrossRef] [Green Version]
- Pereverzev, M.O.; Vygodina, T.V.; Konstantinov, A.A.; Skulachev, V.P. Cytochrome c, an ideal antioxidant. Biochem. Soc. Trans. 2003, 31, 1312–1315. [Google Scholar] [CrossRef] [PubMed]
- Atlante, A.; Calissano, P.; Bobba, A.; Azzariti, A.; Marra, E.; Passarella, S. Cytochrome c is released from mitochondria in a reactive oxygen species (ROS)-dependent fashion and can operate as a ROS scavenger and as a respiratory substrate in cerebellar neurons undergoing excitotoxic death. J. Biol. Chem. 2000, 275, 37159–37166. [Google Scholar] [CrossRef] [Green Version]
- Butler, J.; Jayson, G.G.; Swallow, A.J. The reaction between the superoxide anion radical and cytochrome c. Biochim. Biophys. Acta 1975, 408, 215–222. [Google Scholar] [CrossRef]
- Sharma, R.; Yang, Y.; Sharma, A.; Awasthi, S.; Awasthi, Y.C. Antioxidant role of glutathione S-transferases: Protection against oxidant toxicity and regulation of stress-mediated apoptosis. Antioxid. Redox Signal. 2004, 6, 289–300. [Google Scholar] [CrossRef] [PubMed]
- Perkins, A.; Nelson, K.J.; Parsonage, D.; Poole, L.B.; Karplus, P.A. Peroxiredoxins: Guardians against oxidative stress and modulators of peroxide signaling. Trends Biochem. Sci. 2015, 40, 435–445. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Kim, S.M.; Lee, R.T. Thioredoxin and thioredoxin target proteins: From molecular mechanisms to functional significance. Antioxid. Redox Signal. 2013, 18, 1165–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ursini, F.; Maiorino, M.; Gregolin, C. The selenoenzyme phospholipid hydroperoxide glutathione peroxidase. Biochim. Biophys. Acta 1985, 839, 62–70. [Google Scholar] [CrossRef]
- Flohe, L. The glutathione peroxidase reaction: Molecular basis of the antioxidant function of selenium in mammals. Curr. Top. Cell. Regul. 1985, 27, 473–478. [Google Scholar] [CrossRef]
- Baud, O.; Greene, A.E.; Li, J.; Wang, H.; Volpe, J.J.; Rosenberg, P.A. Glutathione peroxidase-catalase cooperativity is required for resistance to hydrogen peroxide by mature rat oligodendrocytes. J. Neurosci. 2004, 24, 1531–1540. [Google Scholar] [CrossRef] [Green Version]
- Ighodaro, O.M.; Akinloye, O.A. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alex. J. Med. 2018, 54, 287–293. [Google Scholar] [CrossRef] [Green Version]
- McCord, J.M.; Fridovich, I. Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J. Biol. Chem. 1969, 244, 6049–6055. [Google Scholar] [CrossRef]
- Rhee, S.G. Overview on Peroxiredoxin. Mol. Cells 2016, 39, 1–5. [Google Scholar] [CrossRef] [Green Version]
- Arthur, J.R. The glutathione peroxidases. Cell. Mol. Life Sci. 2000, 57, 1825–1835. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Fukai, T.; Ushio-Fukai, M. Superoxide dismutases: Role in redox signaling, vascular function, and diseases. Antioxid. Redox Signal. 2011, 15, 1583–1606. [Google Scholar] [CrossRef] [Green Version]
- Rizvi, S.; Raza, S.T.; Ahmed, F.; Ahmad, A.; Abbas, S.; Mahdi, F. The role of vitamin e in human health and some diseases. Sultan. Qaboos. Univ. Med. J. 2014, 14, e157–e165. [Google Scholar] [PubMed]
- Frei, B. Reactive oxygen species and antioxidant vitamins: Mechanisms of action. Am. J. Med. 1994, 97, 5S–13S. [Google Scholar] [CrossRef]
- Sharp, P.; Hand, D.; Guthrie, E. Quantitative Determination of Dissolved Oxygen: Ascorbic Acid Oxidase Method. Ind. Eng. Chem. Anal. Ed. 1941, 13, 593–597. [Google Scholar] [CrossRef]
- Aldini, G.; Altomare, A.; Baron, G.; Vistoli, G.; Carini, M.; Borsani, L.; Sergio, F. N-Acetylcysteine as an antioxidant and disulphide breaking agent: The reasons why. Free Radic. Res. 2018, 52, 751–762. [Google Scholar] [CrossRef]
- Laurindo, F.R.M. Chapter 10—Redox Cellular Signaling Pathways in Endothelial Dysfunction and Vascular Disease. In Endothelium and Cardiovascular Diseases; Da Luz, P.L., Libby, P., Chagas, A.C.P., Laurindo, F.R.M., Eds.; Academic Press: Cambridge, MA, USA, 2018; pp. 127–145. [Google Scholar]
- Bienert, G.P.; Chaumont, F. Aquaporin-facilitated transmembrane diffusion of hydrogen peroxide. Biochim. Biophys. Acta 2014, 1840, 1596–1604. [Google Scholar] [CrossRef]
- Lennicke, C.; Rahn, J.; Lichtenfels, R.; Wessjohann, L.A.; Seliger, B. Hydrogen peroxide—Production, fate and role in redox signaling of tumor cells. Cell Commun. Signal. 2015, 13, 39. [Google Scholar] [CrossRef] [Green Version]
- Prata, C.; Hrelia, S.; Fiorentini, D. Peroxiporins in Cancer. Int. J. Mol. Sci. 2019, 20, 1371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodrigues, C.; Mósca, A.F.; Martins, A.P.; Nobre, T.; Prista, C.; Antunes, F.; Cipak Gasparovic, A.; Soveral, G. Rat Aquaporin-5 Is pH-Gated Induced by Phosphorylation and Is Implicated in Oxidative Stress. Int. J. Mol. Sci. 2016, 17, 2090. [Google Scholar] [CrossRef] [Green Version]
- Veal, E.A.; Day, A.M.; Morgan, B.A. Hydrogen peroxide sensing and signaling. Mol. Cell 2007, 26, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Schmidt, H.H.; Stocker, R.; Vollbracht, C.; Paulsen, G.; Riley, D.; Daiber, A.; Cuadrado, A. Antioxidants in Translational Medicine. Antioxid. Redox Signal. 2015, 23, 1130–1143. [Google Scholar] [CrossRef] [Green Version]
- Casas, A.I.; Nogales, C.; Mucke, H.A.M.; Petraina, A.; Cuadrado, A.; Rojo, A.I.; Ghezzi, P.; Jaquet, V.; Augsburger, F.; Dufrasne, F.; et al. On the Clinical Pharmacology of Reactive Oxygen Species. Pharm. Rev. 2020, 72, 801–828. [Google Scholar] [CrossRef]
- Kitagishi, Y.; Matsuda, S. Redox regulation of tumor suppressor PTEN in cancer and aging (Review). Int. J. Mol. Med. 2013, 31, 511–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [Green Version]
- Lingappan, K. NF-κB in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef] [PubMed]
- Milkovic, L.; Zarkovic, N.; Saso, L. Controversy about pharmacological modulation of Nrf2 for cancer therapy. Redox Biol. 2017, 12, 727–732. [Google Scholar] [CrossRef] [Green Version]
- 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. [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] [PubMed] [Green Version]
- Filomeni, G.; De Zio, D.; Cecconi, F. Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ. 2015, 22, 377–388. [Google Scholar] [CrossRef] [Green Version]
- Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys. Acta 2016, 1863, 2977–2992. [Google Scholar] [CrossRef] [PubMed]
- Mishra, M.; Duraisamy, A.J.; Bhattacharjee, S.; Kowluru, R.A. Adaptor Protein p66Shc: A Link between Cytosolic and Mitochondrial Dysfunction in the Development of Diabetic Retinopathy. Antioxid. Redox Signal. 2019, 30, 1621–1634. [Google Scholar] [CrossRef] [PubMed]
- Szocs, K. Endothelial dysfunction and reactive oxygen species production in ischemia/reperfusion and nitrate tolerance. Gen. Physiol. Biophys. 2004, 23, 265–295. [Google Scholar] [PubMed]
- Al Sabaani, N. Exendin-4 inhibits high glucose-induced oxidative stress in retinal pigment epithelial cells by modulating the expression and activation of p(66)Shc. Cutan. Ocul. Toxicol. 2021, 40, 175–186. [Google Scholar] [CrossRef]
- Mansouri, A.; Gattolliat, C.H.; Asselah, T. Mitochondrial Dysfunction and Signaling in Chronic Liver Diseases. Gastroenterology 2018, 155, 629–647. [Google Scholar] [CrossRef] [Green Version]
- Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc. Pharm. 2018, 100, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Barnes, P.J. Oxidative stress-based therapeutics in COPD. Redox. Biol. 2020, 33, 101544. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, W.; Ijaz, B.; Shabbiri, K.; Ahmed, F.; Rehman, S. Oxidative toxicity in diabetes and Alzheimer’s disease: Mechanisms behind ROS/ RNS generation. J. Biomed. Sci. 2017, 24, 76. [Google Scholar] [CrossRef]
- Lone, A.; Harris, R.A.; Singh, O.; Betts, D.H.; Cumming, R.C. p66Shc activation promotes increased oxidative phosphorylation and renders CNS cells more vulnerable to amyloid beta toxicity. Sci. Rep. 2018, 8, 17081. [Google Scholar] [CrossRef]
- Irazabal, M.V.; Torres, V.E. Reactive Oxygen Species and Redox Signaling in Chronic Kidney Disease. Cells 2020, 9, 1342. [Google Scholar] [CrossRef]
- Nagar, H.; Piao, S.; Kim, C.S. Role of Mitochondrial Oxidative Stress in Sepsis. Acute Crit. Care 2018, 33, 65–72. [Google Scholar] [CrossRef]
- Kaneto, H.; Katakami, N.; Matsuhisa, M.; Matsuoka, T.A. Role of reactive oxygen species in the progression of type 2 diabetes and atherosclerosis. Mediat. Inflamm. 2010, 2010, 453892. [Google Scholar] [CrossRef] [Green Version]
- Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef] [PubMed]
- Sugamura, K.; Keaney, J.F., Jr. Reactive oxygen species in cardiovascular disease. Free Radic. Biol. Med. 2011, 51, 978–992. [Google Scholar] [CrossRef] [Green Version]
- Daiber, A.; Di Lisa, F.; Ferdinandy, P. Pharmacology of oxidative stress: Translational opportunities. Br. J. Pharm. 2017, 174, 1511–1513. [Google Scholar] [CrossRef] [Green Version]
- Kornfeld, O.S.; Hwang, S.; Disatnik, M.H.; Chen, C.H.; Qvit, N.; Mochly-Rosen, D. Mitochondrial reactive oxygen species at the heart of the matter: New therapeutic approaches for cardiovascular diseases. Circ. Res. 2015, 116, 1783–1799. [Google Scholar] [CrossRef] [PubMed]
- Steinhubl, S.R. Why have antioxidants failed in clinical trials? Am. J. Cardiol. 2008, 101, 14d–19d. [Google Scholar] [CrossRef]
- Pawson, T.; Gish, G.D. SH2 and SH3 domains: From structure to function. Cell 1992, 71, 359–362. [Google Scholar] [CrossRef]
- Kumar, S. P66Shc and vascular endothelial function. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Migliaccio, E.; Mele, S.; Salcini, A.E.; Pelicci, G.; Lai, K.M.; Superti-Furga, G.; Pawson, T.; Di Fiore, P.P.; Lanfrancone, L.; Pelicci, P.G. Opposite effects of the p52shc/p46shc and p66shc splicing isoforms on the EGF receptor-MAP kinase-fos signalling pathway. EMBO J. 1997, 16, 706–716. [Google Scholar] [CrossRef]
- Heusch, G. Mitochondria at the heart of cardiovascular protection: p66shc-friend or foe? Eur. Heart J. 2015, 36, 469–471. [Google Scholar] [CrossRef] [Green Version]
- Akhmedov, A.; Montecucco, F.; Braunersreuther, V.; Camici, G.G.; Jakob, P.; Reiner, M.F.; Glanzmann, M.; Burger, F.; Paneni, F.; Galan, K.; et al. Genetic deletion of the adaptor protein p66Shc increases susceptibility to short-term ischaemic myocardial injury via intracellular salvage pathways. Eur. Heart J. 2015, 36, 516–526a. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giorgio, M.; Berry, A.; Berniakovich, I.; Poletaeva, I.; Trinei, M.; Stendardo, M.; Hagopian, K.; Ramsey, J.J.; Cortopassi, G.; Migliaccio, E.; et al. The p66Shc knocked out mice are short lived under natural condition. Aging Cell 2012, 11, 162–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramsey, J.J.; Tran, D.; Giorgio, M.; Griffey, S.M.; Koehne, A.; Laing, S.T.; Taylor, S.L.; Kim, K.; Cortopassi, G.A.; Lloyd, K.C.; et al. The influence of Shc proteins on life span in mice. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 1177–1185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, K.M.; Pawson, T. The ShcA phosphotyrosine docking protein sensitizes cardiovascular signaling in the mouse embryo. Genes Dev. 2000, 14, 1132–1145. [Google Scholar] [CrossRef] [PubMed]
- Tomilov, A.A.; Ramsey, J.J.; Hagopian, K.; Giorgio, M.; Kim, K.M.; Lam, A.; Migliaccio, E.; Lloyd, K.C.; Berniakovich, I.; Prolla, T.A.; et al. The Shc locus regulates insulin signaling and adiposity in mammals. Aging Cell 2011, 10, 55–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berniakovich, I.; Trinei, M.; Stendardo, M.; Migliaccio, E.; Minucci, S.; Bernardi, P.; Pelicci, P.G.; Giorgio, M. p66Shc-generated oxidative signal promotes fat accumulation. J. Biol. Chem. 2008, 283, 34283–34293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menini, S.; Amadio, L.; Oddi, G.; Ricci, C.; Pesce, C.; Pugliese, F.; Giorgio, M.; Migliaccio, E.; Pelicci, P.; Iacobini, C.; et al. Deletion of p66Shc longevity gene protects against experimental diabetic glomerulopathy by preventing diabetes-induced oxidative stress. Diabetes 2006, 55, 1642–1650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fadini, G.P.; Ceolotto, G.; Pagnin, E.; de Kreutzenberg, S.; Avogaro, A. At the crossroads of longevity and metabolism: The metabolic syndrome and lifespan determinant pathways. Aging Cell 2011, 10, 10–17. [Google Scholar] [CrossRef] [PubMed]
- Zaccagnini, G.; Martelli, F.; Fasanaro, P.; Magenta, A.; Gaetano, C.; Di Carlo, A.; Biglioli, P.; Giorgio, M.; Martin-Padura, I.; Pelicci, P.G.; et al. p66ShcA modulates tissue response to hindlimb ischemia. Circulation 2004, 109, 2917–2923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carpi, A.; Menabo, R.; Kaludercic, N.; Pelicci, P.; Di Lisa, F.; Giorgio, M. The cardioprotective effects elicited by p66(Shc) ablation demonstrate the crucial role of mitochondrial ROS formation in ischemia/reperfusion injury. Biochim. Biophys. Acta 2009, 1787, 774–780. [Google Scholar] [CrossRef] [PubMed]
- Napoli, C.; Martin-Padura, I.; de Nigris, F.; Giorgio, M.; Mansueto, G.; Somma, P.; Condorelli, M.; Sica, G.; De Rosa, G.; Pelicci, P. Deletion of the p66Shc longevity gene reduces systemic and tissue oxidative stress, vascular cell apoptosis, and early atherogenesis in mice fed a high-fat diet. Proc. Natl. Acad. Sci. USA 2003, 100, 2112–2116. [Google Scholar] [CrossRef] [Green Version]
- Graiani, G.; Lagrasta, C.; Migliaccio, E.; Spillmann, F.; Meloni, M.; Madeddu, P.; Quaini, F.; Padura, I.M.; Lanfrancone, L.; Pelicci, P.; et al. Genetic deletion of the p66Shc adaptor protein protects from angiotensin II-induced myocardial damage. Hypertension 2005, 46, 433–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ranieri Sofia, C.; Fusco, S.; Panieri, E.; Labate, V.; Mele, M.; Tesori, V.; Ferrara Anna, M.; Maulucci, G.; De Spirito, M.; Martorana Giuseppe, E.; et al. Mammalian life-span determinant p66shcA mediates obesity-induced insulin resistance. Proc. Natl. Acad. Sci. USA 2010, 107, 13420–13425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stevenson, L.E.; Frackelton, A.R., Jr. Constitutively tyrosine phosphorylated p52 Shc in breast cancer cells: Correlation with ErbB2 and p66 Shc expression. Breast Cancer Res. Treat 1998, 49, 119–128. [Google Scholar] [CrossRef]
- Gertz, M.; Fischer, F.; Wolters, D.; Steegborn, C. Activation of the lifespan regulator p66Shc through reversible disulfide bond formation. Proc. Natl. Acad. Sci. USA 2008, 105, 5705–5709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pelicci, G.; Lanfrancone, L.; Grignani, F.; McGlade, J.; Cavallo, F.; Forni, G.; Nicoletti, I.; Grignani, F.; Pawson, T.; Pelicci, P.G. A novel transforming protein (SHC) with an SH2 domain is implicated in mitogenic signal transduction. Cell 1992, 70, 93–104. [Google Scholar] [CrossRef]
- Berry, A.; Capone, F.; Giorgio, M.; Pelicci, P.G.; de Kloet, E.R.; Alleva, E.; Minghetti, L.; Cirulli, F. Deletion of the life span determinant p66Shc prevents age-dependent increases in emotionality and pain sensitivity in mice. Exp. Gerontol. 2007, 42, 37–45. [Google Scholar] [CrossRef] [PubMed]
- Berry, A.; Greco, A.; Giorgio, M.; Pelicci, P.G.; de Kloet, R.; Alleva, E.; Minghetti, L.; Cirulli, F. Deletion of the lifespan determinant p66(Shc) improves performance in a spatial memory task, decreases levels of oxidative stress markers in the hippocampus and increases levels of the neurotrophin BDNF in adult mice. Exp. Gerontol. 2008, 43, 200–208. [Google Scholar] [CrossRef]
- Conti, L.; De Fraja, C.; Gulisano, M.; Migliaccio, E.; Govoni, S.; Cattaneo, E. Expression and activation of SH2/PTB-containing ShcA adaptor protein reflects the pattern of neurogenesis in the mammalian brain. Proc. Natl. Acad. Sci. USA 1997, 94, 8185–8190. [Google Scholar] [CrossRef] [Green Version]
- Lebiedzinska, M.; Duszynski, J.; Rizzuto, R.; Pinton, P.; Wieckowski, M.R. Age-related changes in levels of p66Shc and serine 36-phosphorylated p66Shc in organs and mouse tissues. Arch. Biochem. Biophys. 2009, 486, 73–80. [Google Scholar] [CrossRef]
- Ventura, A.; Luzi, L.; Pacini, S.; Baldari, C.T.; Pelicci, P.G. The p66Shc longevity gene is silenced through epigenetic modifications of an alternative promoter. J. Biol. Chem. 2002, 277, 22370–22376. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.R.; Kim, C.S.; Naqvi, A.; Kumar, A.; Kumar, S.; Hoffman, T.A.; Irani, K. Epigenetic upregulation of p66shc mediates low-density lipoprotein cholesterol-induced endothelial cell dysfunction. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H189–H196. [Google Scholar] [CrossRef] [Green Version]
- Kim, C.S.; Jung, S.B.; Naqvi, A.; Hoffman, T.A.; DeRicco, J.; Yamamori, T.; Cole, M.P.; Jeon, B.H.; Irani, K. p53 impairs endothelium-dependent vasomotor function through transcriptional upregulation of p66shc. Circ. Res. 2008, 103, 1441–1450. [Google Scholar] [CrossRef]
- Ihling, C.; Haendeler, J.; Menzel, G.; Hess, R.D.; Fraedrich, G.; Schaefer, H.E.; Zeiher, A.M. Co-expression of p53 and MDM2 in human atherosclerosis: Implications for the regulation of cellularity of atherosclerotic lesions. J. Pathol. 1998, 185, 303–312. [Google Scholar] [CrossRef]
- Trinei, M.; Giorgio, M.; Cicalese, A.; Barozzi, S.; Ventura, A.; Migliaccio, E.; Milia, E.; Padura, I.M.; Raker, V.A.; Maccarana, M.; et al. A p53-p66Shc signalling pathway controls intracellular redox status, levels of oxidation-damaged DNA and oxidative stress-induced apoptosis. Oncogene 2002, 21, 3872–3878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed]
- Paneni, F.; Mocharla, P.; Akhmedov, A.; Costantino, S.; Osto, E.; Volpe, M.; Luscher, T.F.; Cosentino, F. Gene silencing of the mitochondrial adaptor p66(Shc) suppresses vascular hyperglycemic memory in diabetes. Circ. Res. 2012, 111, 278–289. [Google Scholar] [CrossRef] [Green Version]
- Costantino, S.; Paneni, F.; Virdis, A.; Hussain, S.; Mohammed, S.A.; Capretti, G.; Akhmedov, A.; Dalgaard, K.; Chiandotto, S.; Pospisilik, J.A.; et al. Interplay among H3K9-editing enzymes SUV39H1, JMJD2C and SRC-1 drives p66Shc transcription and vascular oxidative stress in obesity. Eur. Heart J. 2019, 40, 383–391. [Google Scholar] [CrossRef] [Green Version]
- Bosch-Presegué, L.; Raurell-Vila, H.; Marazuela-Duque, A.; Kane-Goldsmith, N.; Valle, A.; Oliver, J.; Serrano, L.; Vaquero, A. Stabilization of Suv39H1 by SirT1 is part of oxidative stress response and ensures genome protection. Mol. Cell 2011, 42, 210–223. [Google Scholar] [CrossRef]
- Du, W.; Jiang, Y.; Zheng, Z.; Zhang, Z.; Chen, N.; Ma, Z.; Yao, Z.; Terada, L.; Liu, Z. Feedback loop between p66(Shc) and Nrf2 promotes lung cancer progression. Cancer Lett. 2013, 337, 58–65. [Google Scholar] [CrossRef] [PubMed]
- Miyazawa, M.; Tsuji, Y. Evidence for a novel antioxidant function and isoform-specific regulation of the human p66Shc gene. Mol. Biol. Cell 2014, 25, 2116–2127. [Google Scholar] [CrossRef]
- Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal 2012, 24, 981–990. [Google Scholar] [CrossRef] [Green Version]
- Faisal, A.; el-Shemerly, M.; Hess, D.; Nagamine, Y. Serine/threonine phosphorylation of ShcA. Regulation of protein-tyrosine phosphatase-pest binding and involvement in insulin signaling. J. Biol. Chem. 2002, 277, 30144–30152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nemoto, S.; Combs, C.A.; French, S.; Ahn, B.H.; Fergusson, M.M.; Balaban, R.S.; Finkel, T. The mammalian longevity-associated gene product p66shc regulates mitochondrial metabolism. J. Biol. Chem. 2006, 281, 10555–10560. [Google Scholar] [CrossRef] [Green Version]
- Ahmed, S.B.M.; Prigent, S.A. Insights into the Shc Family of Adaptor Proteins. J. Mol. Signal. 2017, 12, 2. [Google Scholar] [CrossRef] [Green Version]
- Ventura, A.; Maccarana, M.; Raker, V.A.; Pelicci, P.G. A cryptic targeting signal induces isoform-specific localization of p46Shc to mitochondria. J. Biol. Chem. 2004, 279, 2299–2306. [Google Scholar] [CrossRef] [Green Version]
- Orsini, F.; Migliaccio, E.; Moroni, M.; Contursi, C.; Raker, V.A.; Piccini, D.; Martin-Padura, I.; Pelliccia, G.; Trinei, M.; Bono, M.; et al. The life span determinant p66Shc localizes to mitochondria where it associates with mitochondrial heat shock protein 70 and regulates trans-membrane potential. J. Biol. Chem. 2004, 279, 25689–25695. [Google Scholar] [CrossRef] [Green Version]
- Gertz, M.; Fischer, F.; Leipelt, M.; Wolters, D.; Steegborn, C. Identification of Peroxiredoxin 1 as a novel interaction partner for the lifespan regulator protein p66Shc. Aging (Albany NY) 2009, 1, 254–265. [Google Scholar] [CrossRef] [PubMed] [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]
- Bhat, S.S.; Anand, D.; Khanday, F.A. p66Shc as a switch in bringing about contrasting responses in cell growth: Implications on cell proliferation and apoptosis. Mol. Cancer 2015, 14, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okada, S.; Kao, A.W.; Ceresa, B.P.; Blaikie, P.; Margolis, B.; Pessin, J.E. The 66-kDa Shc isoform is a negative regulator of the epidermal growth factor-stimulated mitogen-activated protein kinase pathway. J. Biol. Chem. 1997, 272, 28042–28049. [Google Scholar] [CrossRef] [Green Version]
- Pacini, S.; Pellegrini, M.; Migliaccio, E.; Patrussi, L.; Ulivieri, C.; Ventura, A.; Carraro, F.; Naldini, A.; Lanfrancone, L.; Pelicci, P.; et al. p66SHC promotes apoptosis and antagonizes mitogenic signaling in T cells. Mol. Cell. Biol. 2004, 24, 1747–1757. [Google Scholar] [CrossRef] [Green Version]
- Ong, S.H.; Dilworth, S.; Hauck-Schmalenberger, I.; Pawson, T.; Kiefer, F. ShcA and Grb2 mediate polyoma middle T antigen-induced endothelial transformation and Gab1 tyrosine phosphorylation. EMBO J. 2001, 20, 6327–6336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arany, I.; Faisal, A.; Nagamine, Y.; Safirstein, R.L. p66shc inhibits pro-survival epidermal growth factor receptor/ERK signaling during severe oxidative stress in mouse renal proximal tubule cells. J. Biol. Chem. 2008, 283, 6110–6117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xi, G.; Shen, X.; Clemmons, D.R. p66shc negatively regulates insulin-like growth factor I signal transduction via inhibition of p52shc binding to Src homology 2 domain-containing protein tyrosine phosphatase substrate-1 leading to impaired growth factor receptor-bound protein-2 membrane recruitment. Mol. Endocrinol. 2008, 22, 2162–2175. [Google Scholar] [CrossRef] [Green Version]
- Khanday, F.A.; Santhanam, L.; Kasuno, K.; Yamamori, T.; Naqvi, A.; Dericco, J.; Bugayenko, A.; Mattagajasingh, I.; Disanza, A.; Scita, G.; et al. Sos-mediated activation of rac1 by p66shc. J. Cell Biol. 2006, 172, 817–822. [Google Scholar] [CrossRef] [Green Version]
- Khanday, F.A.; Yamamori, T.; Mattagajasingh, I.; Zhang, Z.; Bugayenko, A.; Naqvi, A.; Santhanam, L.; Nabi, N.; Kasuno, K.; Day, B.W.; et al. Rac1 leads to phosphorylation-dependent increase in stability of the p66shc adaptor protein: Role in Rac1-induced oxidative stress. Mol. Biol. Cell. 2006, 17, 122–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qin, F.; Simeone, M.; Patel, R. Inhibition of NADPH oxidase reduces myocardial oxidative stress and apoptosis and improves cardiac function in heart failure after myocardial infarction. Free Radic. Biol. Med. 2007, 43, 271–281. [Google Scholar] [CrossRef]
- Davidson, D.; Veillette, A. PTP-PEST, a scaffold protein tyrosine phosphatase, negatively regulates lymphocyte activation by targeting a unique set of substrates. EMBO J. 2001, 20, 3414–3426. [Google Scholar] [CrossRef]
- Schneider, E.; Keppler, R.; Prawitt, D.; Steinwender, C.; Roos, F.C.; Thuroff, J.W.; Lausch, E.; Brenner, W. Migration of renal tumor cells depends on dephosphorylation of Shc by PTEN. Int. J. Oncol. 2011, 38, 823–831. [Google Scholar] [CrossRef] [Green Version]
- Kraut-Cohen, J.; Muller, W.J.; Elson, A. Protein-tyrosine phosphatase epsilon regulates Shc signaling in a kinase-specific manner: Increasing coherence in tyrosine phosphatase signaling. J. Biol. Chem. 2008, 283, 4612–4621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vikram, A.; Kim, Y.R.; Kumar, S.; Naqvi, A.; Hoffman, T.A.; Kumar, A.; Miller, F.J., Jr.; Kim, C.S.; Irani, K. Canonical Wnt signaling induces vascular endothelial dysfunction via p66Shc-regulated reactive oxygen species. Arter. Thromb. Vasc. Biol. 2014, 34, 2301–2309. [Google Scholar] [CrossRef] [Green Version]
- Di Lisa, F.; Giorgio, M.; Ferdinandy, P.; Schulz, R. New aspects of p66Shc in ischaemia reperfusion injury and other cardiovascular diseases. Br. J. Pharm. 2017, 174, 1690–1703. [Google Scholar] [CrossRef] [Green Version]
- Shi, Y.; Cosentino, F.; Camici, G.G.; Akhmedov, A.; Vanhoutte, P.M.; Tanner, F.C.; Luscher, T.F. Oxidized low-density lipoprotein activates p66Shc via lectin-like oxidized low-density lipoprotein receptor-1, protein kinase C-beta, and c-Jun N-terminal kinase kinase in human endothelial cells. Arter. Thromb. Vasc. Biol. 2011, 31, 2090–2097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pinton, P.; Rimessi, A.; Marchi, S.; Orsini, F.; Migliaccio, E.; Giorgio, M.; Contursi, C.; Minucci, S.; Mantovani, F.; Wieckowski, M.R.; et al. Protein kinase C beta and prolyl isomerase 1 regulate mitochondrial effects of the life-span determinant p66Shc. Science 2007, 315, 659–663. [Google Scholar] [CrossRef] [PubMed]
- Xie, Z.Z.; Shi, M.M.; Xie, L.; Wu, Z.Y.; Li, G.; Hua, F.; Bian, J.S. Sulfhydration of p66Shc at cysteine59 mediates the antioxidant effect of hydrogen sulfide. Antioxid. Redox Signal. 2014, 21, 2531–2542. [Google Scholar] [CrossRef] [PubMed]
- 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] [Green Version]
- Wu, R.F.; Liao, C.; Fu, G.; Hayenga, H.N.; Yang, K.; Ma, Z.; Liu, Z.; Terada, L.S. p66(Shc) Couples Mechanical Signals to RhoA through Focal Adhesion Kinase-Dependent Recruitment of p115-RhoGEF and GEF-H1. Mol. Cell. Biol. 2016, 36, 2824–2837. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.; Myers, D.P.; Wu, R.F.; Nwariaku, F.E.; Terada, L.S. p66Shc mediates anoikis through RhoA. J. Cell Biol. 2007, 179, 23–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, K.D.; Li, Y.S.; Kim, M.; Li, S.; Yuan, S.; Chien, S.; Shyy, J.Y. Mechanotransduction in response to shear stress. Roles of receptor tyrosine kinases, integrins, and Shc. J. Biol. Chem. 1999, 274, 18393–18400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wary, K.K.; Mainiero, F.; Isakoff, S.J.; Marcantonio, E.E.; Giancotti, F.G. The adaptor protein Shc couples a class of integrins to the control of cell cycle progression. Cell 1996, 87, 733–743. [Google Scholar] [CrossRef] [Green Version]
- Vanderlaan, R.D.; Hardy, W.R.; Kabir, M.G.; Pasculescu, A.; Jones, N.; deTombe, P.P.; Backx, P.H.; Pawson, T. The ShcA phosphotyrosine docking protein uses distinct mechanisms to regulate myocyte and global heart function. Circ. Res. 2011, 108, 184–193. [Google Scholar] [CrossRef] [Green Version]
- Hardy, W.R.; Li, L.; Wang, Z.; Sedy, J.; Fawcett, J.; Frank, E.; Kucera, J.; Pawson, T. Combinatorial ShcA docking interactions support diversity in tissue morphogenesis. Science 2007, 317, 251–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Natalicchio, A.; Laviola, L.; De Tullio, C.; Renna, L.A.; Montrone, C.; Perrini, S.; Valenti, G.; Procino, G.; Svelto, M.; Giorgino, F. Role of the p66Shc isoform in insulin-like growth factor I receptor signaling through MEK/Erk and regulation of actin cytoskeleton in rat myoblasts. J. Biol. Chem. 2004, 279, 43900–43909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Favetta, L.A.; Madan, P.; Mastromonaco, G.F.; St John, E.J.; King, W.A.; Betts, D.H. The oxidative stress adaptor p66Shc is required for permanent embryo arrest in vitro. BMC Dev. Biol. 2007, 7, 132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, M.K.; Smith, S.M.; Banerjee, M.M.; Li, C.; Minoo, P.; Volpe, M.V.; Nielsen, H.C. The p66Shc adapter protein regulates the morphogenesis and epithelial maturation of fetal mouse lungs. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 306, L316–L325. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.; Liu, Z.; Wu, R.F.; Terada, L.S. p66(Shc) restrains Ras hyperactivation and suppresses metastatic behavior. Oncogene 2010, 29, 5559–5567. [Google Scholar] [CrossRef] [Green Version]
- Dohn, M.R.; Brown, M.V.; Reynolds, A.B. An essential role for p120-catenin in Src- and Rac1-mediated anchorage-independent cell growth. J. Cell Biol. 2009, 184, 437–450. [Google Scholar] [CrossRef] [Green Version]
- Dupont, S.; Morsut, L.; Aragona, M.; Enzo, E.; Giulitti, S.; Cordenonsi, M.; Zanconato, F.; Le Digabel, J.; Forcato, M.; Bicciato, S.; et al. Role of YAP/TAZ in mechanotransduction. Nature 2011, 474, 179–183. [Google Scholar] [CrossRef] [PubMed]
- Terada, L.S. Shc and the mechanotransduction of cellular anchorage and metastasis. Small GTPases 2019, 10, 64–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dutta, P.; Courties, G.; Wei, Y.; Leuschner, F.; Gorbatov, R.; Robbins, C.S.; Iwamoto, Y.; Thompson, B.; Carlson, A.L.; Heidt, T.; et al. Myocardial infarction accelerates atherosclerosis. Nature 2012, 487, 325–329. [Google Scholar] [CrossRef] [Green Version]
- Martin-Padura, I.; de Nigris, F.; Migliaccio, E.; Mansueto, G.; Minardi, S.; Rienzo, M.; Lerman, L.O.; Stendardo, M.; Giorgio, M.; De Rosa, G.; et al. p66Shc deletion confers vascular protection in advanced atherosclerosis in hypercholesterolemic apolipoprotein E knockout mice. Endothelium 2008, 15, 276–287. [Google Scholar] [CrossRef] [PubMed]
- Haslem, L.; Hays, J.M.; Schmitz, H.; Matsuzaki, S.; Sjoelund, V.; Byrum, S.D.; Humphries, K.M.; Frazer, J.K.; Demeler, B.; Benbrook, D.M.; et al. p66Shc is an apoptotic rheostat whose targeted ROS inhibition improves MI outcomes. bioRxiv 2022, 2022.2004.2014.487897. [Google Scholar] [CrossRef]
- Neumann, C.A.; Cao, J.; Manevich, Y. Peroxiredoxin 1 and its role in cell signaling. Cell Cycle 2009, 8, 4072–4078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirotsu, S.; Abe, Y.; Okada, K.; Nagahara, N.; Hori, H.; Nishino, T.; Hakoshima, T. Crystal structure of a multifunctional 2-Cys peroxiredoxin heme-binding protein 23 kDa/proliferation-associated gene product. Proc. Natl. Acad. Sci. USA 1999, 96, 12333–12338. [Google Scholar] [CrossRef] [Green Version]
- Wood, Z.A.; Schroder, E.; Robin Harris, J.; Poole, L.B. Structure, mechanism and regulation of peroxiredoxins. Trends Biochem. Sci. 2003, 28, 32–40. [Google Scholar] [CrossRef]
- Kim, S.Y.; Kim, T.J.; Lee, K.Y. A novel function of peroxiredoxin 1 (Prx-1) in apoptosis signal-regulating kinase 1 (ASK1)-mediated signaling pathway. FEBS Lett. 2008, 582, 1913–1918. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.J.; Lee, W.S.; Ip, C.; Chae, H.Z.; Park, E.M.; Park, Y.M. Prx1 suppresses radiation-induced c-Jun NH2-terminal kinase signaling in lung cancer cells through interaction with the glutathione S-transferase Pi/c-Jun NH2-terminal kinase complex. Cancer Res. 2006, 66, 7136–7142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, B.; Wang, Y.; Su, Y. Peroxiredoxins, a novel target in cancer radiotherapy. Cancer Lett 2009, 286, 154–160. [Google Scholar] [CrossRef]
- Lee, W.; Choi, K.S.; Riddell, J.; Ip, C.; Ghosh, D.; Park, J.H.; Park, Y.M. Human peroxiredoxin 1 and 2 are not duplicate proteins: The unique presence of CYS83 in Prx1 underscores the structural and functional differences between Prx1 and Prx2. J. Biol. Chem. 2007, 282, 22011–22022. [Google Scholar] [CrossRef]
- Shau, H.; Gupta, R.K.; Golub, S.H. Identification of a natural killer enhancing factor (NKEF) from human erythroid cells. Cell Immunol. 1993, 147, 1–11. [Google Scholar] [CrossRef]
- Rhee, S.G.; Woo, H.A. Multiple functions of peroxiredoxins: Peroxidases, sensors and regulators of the intracellular messenger H2O2, and protein chaperones. Antioxid. Redox Signal. 2011, 15, 781–794. [Google Scholar] [CrossRef] [PubMed]
- Ishii, T.; Warabi, E.; Yanagawa, T. Novel roles of peroxiredoxins in inflammation, cancer and innate immunity. J. Clin. Biochem. Nutr. 2012, 50, 91–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moon, J.C.; Hah, Y.S.; Kim, W.Y.; Jung, B.G.; Jang, H.H.; Lee, J.R.; Kim, S.Y.; Lee, Y.M.; Jeon, M.G.; Kim, C.W.; et al. Oxidative stress-dependent structural and functional switching of a human 2-Cys peroxiredoxin isotype II that enhances HeLa cell resistance to H2O2-induced cell death. J. Biol. Chem. 2005, 280, 28775–28784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wen, S.T.; Van Etten, R.A. The PAG gene product, a stress-induced protein with antioxidant properties, is an Abl SH3-binding protein and a physiological inhibitor of c-Abl tyrosine kinase activity. Genes Dev. 1997, 11, 2456–2467. [Google Scholar] [CrossRef] [Green Version]
- Mu, Z.M.; Yin, X.Y.; Prochownik, E.V. Pag, a putative tumor suppressor, interacts with the Myc Box II domain of c-Myc and selectively alters its biological function and target gene expression. J. Biol. Chem. 2002, 277, 43175–43184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riddell, J.R.; Bshara, W.; Moser, M.T.; Spernyak, J.A.; Foster, B.A.; Gollnick, S.O. Peroxiredoxin 1 controls prostate cancer growth through Toll-like receptor 4-dependent regulation of tumor vasculature. Cancer Res. 2011, 71, 1637–1646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ren, P.; Ye, H.; Dai, L.; Liu, M.; Liu, X.; Chai, Y.; Shao, Q.; Li, Y.; Lei, N.; Peng, B.; et al. Peroxiredoxin 1 is a tumor-associated antigen in esophageal squamous cell carcinoma. Oncol. Rep. 2013, 30, 2297–2303. [Google Scholar] [CrossRef] [Green Version]
- Karihtala, P.; Mantyniemi, A.; Kang, S.W.; Kinnula, V.L.; Soini, Y. Peroxiredoxins in breast carcinoma. Clin. Cancer Res. 2003, 9, 3418–3424. [Google Scholar] [PubMed]
- Deng, B.; Ye, N.; Luo, G.; Chen, X.; Wang, Y. Proteomics analysis of stage-specific proteins expressed in human squamous cell lung carcinoma tissues. Cancer Biomark 2005, 1, 279–286. [Google Scholar] [CrossRef]
- Jang, H.H.; Lee, K.O.; Chi, Y.H.; Jung, B.G.; Park, S.K.; Park, J.H.; Lee, J.R.; Lee, S.S.; Moon, J.C.; Yun, J.W.; et al. Two enzymes in one; two yeast peroxiredoxins display oxidative stress-dependent switching from a peroxidase to a molecular chaperone function. Cell 2004, 117, 625–635. [Google Scholar] [CrossRef]
- Levav-Cohen, Y.; Goldberg, Z.; Zuckerman, V.; Grossman, T.; Haupt, S.; Haupt, Y. C-Abl as a modulator of p53. Biochem. Biophys. Res. Commun. 2005, 331, 737–749. [Google Scholar] [CrossRef]
- Neumann, C.A.; Fang, Q. Are peroxiredoxins tumor suppressors? Curr. Opin. Pharm. 2007, 7, 375–380. [Google Scholar] [CrossRef]
- Hansen, J.M.; Moriarty-Craige, S.; Jones, D.P. Nuclear and cytoplasmic peroxiredoxin-1 differentially regulate NF-kappaB activities. Free Radic. Biol. Med. 2007, 43, 282–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chhipa, R.R.; Lee, K.S.; Onate, S.; Wu, Y.; Ip, C. Prx1 enhances androgen receptor function in prostate cancer cells by increasing receptor affinity to dihydrotestosterone. Mol. Cancer Res. 2009, 7, 1543–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Budanov, A.V. The role of tumor suppressor p53 in the antioxidant defense and metabolism. Subcell Biochem. 2014, 85, 337–358. [Google Scholar] [CrossRef] [Green Version]
- Xiao, L.; Chen, D.; Hu, P.; Wu, J.; Liu, W.; Zhao, Y.; Cao, M.; Fang, Y.; Bi, W.; Zheng, Z.; et al. The c-Abl-MST1 signaling pathway mediates oxidative stress-induced neuronal cell death. J. Neurosci. 2011, 31, 9611–9619. [Google Scholar] [CrossRef]
- Morinaka, A.; Funato, Y.; Uesugi, K.; Miki, H. Oligomeric peroxiredoxin-I is an essential intermediate for p53 to activate MST1 kinase and apoptosis. Oncogene 2011, 30, 4208–4218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Butterworth, P.J. Protein kinase C: Current concepts and future perspectives. Cell Biochem. Funct. 1993, 11, 292. [Google Scholar] [CrossRef]
- Mehta, N.K.; Mehta, K.D. Protein kinase C-beta: An emerging connection between nutrient excess and obesity. Biochim. Biophys. Acta 2014, 1841, 1491–1497. [Google Scholar] [CrossRef]
- Newton, A.C. Protein kinase C: Structure, function, and regulation. J. Biol. Chem. 1995, 270, 28495–28498. [Google Scholar] [CrossRef] [Green Version]
- Nishizuka, Y. Protein kinase C and lipid signaling for sustained cellular responses. FASEB J. 1995, 9, 484–496. [Google Scholar] [CrossRef]
- Ono, Y.; Kikkawa, U.; Ogita, K.; Fujii, T.; Kurokawa, T.; Asaoka, Y.; Sekiguchi, K.; Ase, K.; Igarashi, K.; Nishizuka, Y. Expression and properties of two types of protein kinase C: Alternative splicing from a single gene. Science 1987, 236, 1116–1120. [Google Scholar] [CrossRef]
- Leonard, T.A.; Różycki, B.; Saidi, L.F.; Hummer, G.; Hurley, J.H. Crystal structure and allosteric activation of protein kinase C βII. Cell 2011, 144, 55–66. [Google Scholar] [CrossRef] [Green Version]
- Grodsky, N.; Li, Y.; Bouzida, D.; Love, R.; Jensen, J.; Nodes, B.; Nonomiya, J.; Grant, S. Structure of the catalytic domain of human protein kinase C beta II complexed with a bisindolylmaleimide inhibitor. Biochemistry 2006, 45, 13970–13981. [Google Scholar] [CrossRef]
- Newton, A.C. Regulation of the ABC kinases by phosphorylation: Protein kinase C as a paradigm. Biochem. J. 2003, 370, 361–371. [Google Scholar] [CrossRef]
- Newton, A.C. Lipid activation of protein kinases. J. Lipid Res. 2009, 50, S266–S271. [Google Scholar] [CrossRef] [Green Version]
- Parker, P.J.; Parkinson, S.J. AGC protein kinase phosphorylation and protein kinase C. Biochem. Soc. Trans. 2001, 29, 860–863. [Google Scholar] [CrossRef]
- Konishi, H.; Tanaka, M.; Takemura, Y.; Matsuzaki, H.; Ono, Y.; Kikkawa, U.; Nishizuka, Y. Activation of protein kinase C by tyrosine phosphorylation in response to H2O2. Proc. Natl. Acad. Sci. USA 1997, 94, 11233–11237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cosentino-Gomes, D.; Rocco-Machado, N.; Meyer-Fernandes, J.R. Cell signaling through protein kinase C oxidation and activation. Int. J. Mol. Sci. 2012, 13, 10697–10721. [Google Scholar] [CrossRef] [Green Version]
- Ikenoue, T.; Inoki, K.; Yang, Q.; Zhou, X.; Guan, K.L. Essential function of TORC2 in PKC and Akt turn motif phosphorylation, maturation and signalling. EMBO J. 2008, 27, 1919–1931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarbassov, D.D.; Ali, S.M.; Kim, D.H.; Guertin, D.A.; Latek, R.R.; Erdjument-Bromage, H.; Tempst, P.; Sabatini, D.M. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr. Biol. 2004, 14, 1296–1302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, D.; Gould, C.; Garza, R.; Gao, T.; Hampton, R.Y.; Newton, A.C. Amplitude control of protein kinase C by RINCK, a novel E3 ubiquitin ligase. J. Biol. Chem. 2007, 282, 33776–33787. [Google Scholar] [CrossRef] [Green Version]
- Gould, C.M.; Kannan, N.; Taylor, S.S.; Newton, A.C. The chaperones Hsp90 and Cdc37 mediate the maturation and stabilization of protein kinase C through a conserved PXXP motif in the C-terminal tail. J. Biol. Chem. 2009, 284, 4921–4935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scivittaro, V.; Ganz, M.B.; Weiss, M.F. AGEs induce oxidative stress and activate protein kinase C-beta(II) in neonatal mesangial cells. Am. J. Physiol. Ren. Physiol. 2000, 278, F676–F683. [Google Scholar] [CrossRef] [Green Version]
- Rimessi, A.; Rizzuto, R.; Pinton, P. Differential recruitment of PKC isoforms in HeLa cells during redox stress. Cell Stress Chaperones 2007, 12, 291–298. [Google Scholar] [CrossRef] [PubMed]
- Almeida, M.; Han, L.; Ambrogini, E.; Bartell, S.M.; Manolagas, S.C. Oxidative stress stimulates apoptosis and activates NF-kappaB in osteoblastic cells via a PKCbeta/p66shc signaling cascade: Counter regulation by estrogens or androgens. Mol. Endocrinol. 2010, 24, 2030–2037. [Google Scholar] [CrossRef] [Green Version]
- Gopalakrishna, R.; Jaken, S. Protein kinase C signaling and oxidative stress. Free Radic. Biol. Med. 2000, 28, 1349–1361. [Google Scholar] [CrossRef]
- Giorgi, C.; Agnoletto, C.; Baldini, C.; Bononi, A.; Bonora, M.; Marchi, S.; Missiroli, S.; Patergnani, S.; Poletti, F.; Rimessi, A.; et al. Redox control of protein kinase C: Cell- and disease-specific aspects. Antioxid. Redox Signal. 2010, 13, 1051–1085. [Google Scholar] [CrossRef]
- Choo, H.J.; Kim, J.H.; Kwon, O.B.; Lee, C.S.; Mun, J.Y.; Han, S.S.; Yoon, Y.S.; Yoon, G.; Choi, K.M.; Ko, Y.G. Mitochondria are impaired in the adipocytes of type 2 diabetic mice. Diabetologia 2006, 49, 784–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef]
- Keaney, J.F., Jr.; Larson, M.G.; Vasan, R.S.; Wilson, P.W.; Lipinska, I.; Corey, D.; Massaro, J.M.; Sutherland, P.; Vita, J.A.; Benjamin, E.J.; et al. Obesity and systemic oxidative stress: Clinical correlates of oxidative stress in the Framingham Study. Arter. Thromb. Vasc. Biol. 2003, 23, 434–439. [Google Scholar] [CrossRef] [Green Version]
- Kusminski, C.M.; Scherer, P.E. Mitochondrial dysfunction in white adipose tissue. Trends Endocrinol. Metab. 2012, 23, 435–443. [Google Scholar] [CrossRef] [Green Version]
- Patti, M.E.; Corvera, S. The role of mitochondria in the pathogenesis of type 2 diabetes. Endocr. Rev. 2010, 31, 364–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rong, J.X.; Qiu, Y.; Hansen, M.K.; Zhu, L.; Zhang, V.; Xie, M.; Okamoto, Y.; Mattie, M.D.; Higashiyama, H.; Asano, S.; et al. Adipose mitochondrial biogenesis is suppressed in db/db and high-fat diet-fed mice and improved by rosiglitazone. Diabetes 2007, 56, 1751–1760. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Goldman, S.; Baerga, R.; Zhao, Y.; Komatsu, M.; Jin, S. Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 19860–19865. [Google Scholar] [CrossRef] [Green Version]
- Jansen, H.J.; van Essen, P.; Koenen, T.; Joosten, L.A.; Netea, M.G.; Tack, C.J.; Stienstra, R. Autophagy activity is up-regulated in adipose tissue of obese individuals and modulates proinflammatory cytokine expression. Endocrinology 2012, 153, 5866–5874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nunez, C.E.; Rodrigues, V.S.; Gomes, F.S.; Moura, R.F.; Victorio, S.C.; Bombassaro, B.; Chaim, E.A.; Pareja, J.C.; Geloneze, B.; Velloso, L.A.; et al. Defective regulation of adipose tissue autophagy in obesity. Int. J. Obes. 2013, 37, 1473–1480. [Google Scholar] [CrossRef] [Green Version]
- Green, D.R.; Galluzzi, L.; Kroemer, G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011, 333, 1109–1112. [Google Scholar] [CrossRef] [Green Version]
- Patergnani, S.; Marchi, S.; Rimessi, A.; Bonora, M.; Giorgi, C.; Mehta, K.D.; Pinton, P. PRKCB/protein kinase C, beta and the mitochondrial axis as key regulators of autophagy. Autophagy 2013, 9, 1367–1385. [Google Scholar] [CrossRef] [Green Version]
- Goodyear, L.J.; Kahn, B.B. Exercise, glucose transport, and insulin sensitivity. Annu. Rev. Med. 1998, 49, 235–261. [Google Scholar] [CrossRef]
- Krssak, M.; Falk Petersen, K.; Dresner, A.; DiPietro, L.; Vogel, S.M.; Rothman, D.L.; Roden, M.; Shulman, G.I. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: A 1H NMR spectroscopy study. Diabetologia 1999, 42, 113–116. [Google Scholar] [CrossRef] [Green Version]
- Hennige, A.M.; Heni, M.; Machann, J.; Staiger, H.; Sartorius, T.; Hoene, M.; Lehmann, R.; Weigert, C.; Peter, A.; Bornemann, A.; et al. Enforced expression of protein kinase C in skeletal muscle causes physical inactivity, fatty liver and insulin resistance in the brain. J. Cell. Mol. Med. 2010, 14, 903–913. [Google Scholar] [CrossRef]
- Mir, H.A.; Ali, R.; Mushtaq, U.; Khanday, F.A. Structure-functional implications of longevity protein p66Shc in health and disease. Ageing Res. Rev. 2020, 63, 101139. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.K.; Hammerling, U. The mitochondrial PKCdelta/retinol signal complex exerts real-time control on energy homeostasis. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2020, 1865, 158614. [Google Scholar] [CrossRef]
- Morita, M.; Matsuzaki, H.; Yamamoto, T.; Fukami, Y.; Kikkawa, U. Epidermal growth factor receptor phosphorylates protein kinase C {delta} at Tyr332 to form a trimeric complex with p66Shc in the H2O2-stimulated cells. J. Biochem. 2008, 143, 31–38. [Google Scholar] [CrossRef] [PubMed]
- Acin-Perez, R.; Hoyos, B.; Gong, J.; Vinogradov, V.; Fischman, D.A.; Leitges, M.; Borhan, B.; Starkov, A.; Manfredi, G.; Hammerling, U. Regulation of intermediary metabolism by the PKCdelta signalosome in mitochondria. FASEB J. 2010, 24, 5033–5042. [Google Scholar] [CrossRef] [Green Version]
- Hoyos, B.; Acin-Perez, R.; Fischman, D.A.; Manfredi, G.; Hammerling, U. Hiding in plain sight: Uncovering a new function of vitamin A in redox signaling. Biochim. Biophys. Acta 2012, 1821, 241–247. [Google Scholar] [CrossRef] [PubMed]
- Imam, A.; Hoyos, B.; Swenson, C.; Levi, E.; Chua, R.; Viriya, E.; Hammerling, U. Retinoids as ligands and coactivators of protein kinase C alpha. FASEB J. 2001, 15, 28–30. [Google Scholar] [CrossRef] [PubMed]
- Hoyos, B.; Imam, A.; Chua, R.; Swenson, C.; Tong, G.X.; Levi, E.; Noy, N.; Hämmerling, U. The cysteine-rich regions of the regulatory domains of Raf and protein kinase C as retinoid receptors. J. Exp. Med. 2000, 192, 835–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frank, H.A.; Brudvig, G.W. Redox functions of carotenoids in photosynthesis. Biochemistry 2004, 43, 8607–8615. [Google Scholar] [CrossRef]
- Gong, J.; Hoyos, B.; Acin-Perez, R.; Vinogradov, V.; Shabrova, E.; Zhao, F.; Leitges, M.; Fischman, D.; Manfredi, G.; Hammerling, U. Two protein kinase C isoforms, δ and ε, regulate energy homeostasis in mitochondria by transmitting opposing signals to the pyruvate dehydrogenase complex. FASEB J. 2012, 26, 3537–3549. [Google Scholar] [CrossRef] [Green Version]
- Acin-Perez, R.; Hoyos, B.; Zhao, F.; Vinogradov, V.; Fischman, D.A.; Harris, R.A.; Leitges, M.; Wongsiriroj, N.; Blaner, W.S.; Manfredi, G.; et al. Control of oxidative phosphorylation by vitamin A illuminates a fundamental role in mitochondrial energy homoeostasis. FASEB J. 2010, 24, 627–636. [Google Scholar] [CrossRef] [Green Version]
- Hoyos, B.; Imam, A.; Korichneva, I.; Levi, E.; Chua, R.; Hammerling, U. Activation of c-Raf kinase by ultraviolet light. Regulation by retinoids. J. Biol. Chem. 2002, 277, 23949–23957. [Google Scholar] [CrossRef] [Green Version]
- Mott, H.R.; Carpenter, J.W.; Zhong, S.; Ghosh, S.; Bell, R.M.; Campbell, S.L. The solution structure of the Raf-1 cysteine-rich domain: A novel ras and phospholipid binding site. Proc. Natl. Acad. Sci. USA 1996, 93, 8312–8317. [Google Scholar] [CrossRef] [Green Version]
- Sakai, Y.; Meno, C.; Fujii, H.; Nishino, J.; Shiratori, H.; Saijoh, Y.; Rossant, J.; Hamada, H. The retinoic acid-inactivating enzyme CYP26 is essential for establishing an uneven distribution of retinoic acid along the anterio-posterior axis within the mouse embryo. Genes Dev. 2001, 15, 213–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leitges, M.; Gimborn, K.; Elis, W.; Kalesnikoff, J.; Hughes, M.R.; Krystal, G.; Huber, M. Protein kinase C-delta is a negative regulator of antigen-induced mast cell degranulation. Mol. Cell. Biol. 2002, 22, 3970–3980. [Google Scholar] [CrossRef] [Green Version]
- Bezy, O.; Tran, T.T.; Pihlajamäki, J.; Suzuki, R.; Emanuelli, B.; Winnay, J.; Mori, M.A.; Haas, J.; Biddinger, S.B.; Leitges, M.; et al. PKCδ regulates hepatic insulin sensitivity and hepatosteatosis in mice and humans. J. Clin. Investig. 2011, 121, 2504–2517. [Google Scholar] [CrossRef] [PubMed]
- Schutkowski, M.; Bernhardt, A.; Zhou, X.Z.; Shen, M.; Reimer, U.; Rahfeld, J.U.; Lu, K.P.; Fischer, G. Role of phosphorylation in determining the backbone dynamics of the serine/threonine-proline motif and Pin1 substrate recognition. Biochemistry 1998, 37, 5566–5575. [Google Scholar] [CrossRef] [PubMed]
- Liou, Y.C.; Ryo, A.; Huang, H.K.; Lu, P.J.; Bronson, R.; Fujimori, F.; Uchida, T.; Hunter, T.; Lu, K.P. Loss of Pin1 function in the mouse causes phenotypes resembling cyclin D1-null phenotypes. Proc. Natl. Acad. Sci. USA 2002, 99, 1335–1340. [Google Scholar] [CrossRef] [Green Version]
- Ryo, A.; Nakamura, M.; Wulf, G.; Liou, Y.C.; Lu, K.P. Pin1 regulates turnover and subcellular localization of beta-catenin by inhibiting its interaction with APC. Nat. Cell Biol. 2001, 3, 793–801. [Google Scholar] [CrossRef]
- Wulf, G.M.; Ryo, A.; Wulf, G.G.; Lee, S.W.; Niu, T.; Petkova, V.; Lu, K.P. Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun towards cyclin D1. Embo J. 2001, 20, 3459–3472. [Google Scholar] [CrossRef] [Green Version]
- Ranganathan, R.; Lu, K.P.; Hunter, T.; Noel, J.P. Structural and functional analysis of the mitotic rotamase Pin1 suggests substrate recognition is phosphorylation dependent. Cell 1997, 89, 875–886. [Google Scholar] [CrossRef] [Green Version]
- Lu, K.P.; Liou, Y.C.; Zhou, X.Z. Pinning down proline-directed phosphorylation signaling. Trends Cell Biol. 2002, 12, 164–172. [Google Scholar] [CrossRef]
- Feng, D.; Yao, J.; Wang, G.; Li, Z.; Zu, G.; Li, Y.; Luo, F.; Ning, S.; Qasim, W.; Chen, Z.; et al. Inhibition of p66Shc-mediated mitochondrial apoptosis via targeting prolyl-isomerase Pin1 attenuates intestinal ischemia/reperfusion injury in rats. Clin. Sci. 2017, 131, 759–773. [Google Scholar] [CrossRef]
- Lu, P.J.; Zhou, X.Z.; Liou, Y.C.; Noel, J.P.; Lu, K.P. Critical role of WW domain phosphorylation in regulating phosphoserine binding activity and Pin1 function. J. Biol. Chem. 2002, 277, 2381–2384. [Google Scholar] [CrossRef] [Green Version]
- Wu, X.; Wilcox, C.B.; Devasahayam, G.; Hackett, R.L.; Arévalo-Rodríguez, M.; Cardenas, M.E.; Heitman, J.; Hanes, S.D. The Ess1 prolyl isomerase is linked to chromatin remodeling complexes and the general transcription machinery. Embo J. 2000, 19, 3727–3738. [Google Scholar] [CrossRef] [PubMed]
- Lu, K.P.; Hanes, S.D.; Hunter, T. A human peptidyl-prolyl isomerase essential for regulation of mitosis. Nature 1996, 380, 544–547. [Google Scholar] [CrossRef]
- Hani, J.; Schelbert, B.; Bernhardt, A.; Domdey, H.; Fischer, G.; Wiebauer, K.; Rahfeld, J.U. Mutations in a peptidylprolyl-cis/trans-isomerase gene lead to a defect in 3’-end formation of a pre-mRNA in Saccharomyces cerevisiae. J. Biol. Chem. 1999, 274, 108–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rippmann, J.F.; Hobbie, S.; Daiber, C.; Guilliard, B.; Bauer, M.; Birk, J.; Nar, H.; Garin-Chesa, P.; Rettig, W.J.; Schnapp, A. Phosphorylation-dependent proline isomerization catalyzed by Pin1 is essential for tumor cell survival and entry into mitosis. Cell Growth Differ. 2000, 11, 409–416. [Google Scholar]
- Stukenberg, P.T.; Kirschner, M.W. Pin1 acts catalytically to promote a conformational change in Cdc25. Mol. Cell 2001, 7, 1071–1083. [Google Scholar] [CrossRef]
- Winkler, K.E.; Swenson, K.I.; Kornbluth, S.; Means, A.R. Requirement of the prolyl isomerase Pin1 for the replication checkpoint. Science 2000, 287, 1644–1647. [Google Scholar] [CrossRef]
- Zhou, X.Z.; Kops, O.; Werner, A.; Lu, P.J.; Shen, M.; Stoller, G.; Küllertz, G.; Stark, M.; Fischer, G.; Lu, K.P. Pin1-dependent prolyl isomerization regulates dephosphorylation of Cdc25C and tau proteins. Mol. Cell 2000, 6, 873–883. [Google Scholar] [CrossRef]
- Huang, H.K.; Forsburg, S.L.; John, U.P.; O’Connell, M.J.; Hunter, T. Isolation and characterization of the Pin1/Ess1p homologue in Schizosaccharomyces pombe. J. Cell. Sci. 2001, 114, 3779–3788. [Google Scholar] [CrossRef] [PubMed]
- Price, N.E.; Mumby, M.C. Effects of regulatory subunits on the kinetics of protein phosphatase 2A. Biochemistry 2000, 39, 11312–11318. [Google Scholar] [CrossRef]
- Elgenaidi, I.S.; Spiers, J.P. Regulation of the phosphoprotein phosphatase 2A system and its modulation during oxidative stress: A potential therapeutic target? Pharmacol. Ther. 2019, 198, 68–89. [Google Scholar] [CrossRef]
- Kremmer, E.; Ohst, K.; Kiefer, J.; Brewis, N.; Walter, G. Separation of PP2A core enzyme and holoenzyme with monoclonal antibodies against the regulatory A subunit: Abundant expression of both forms in cells. Mol. Cell. Biol. 1997, 17, 1692–1701. [Google Scholar] [CrossRef] [Green Version]
- Heijman, J.; Dewenter, M.; El-Armouche, A.; Dobrev, D. Function and regulation of serine/threonine phosphatases in the healthy and diseased heart. J. Mol. Cell. Cardiol. 2013, 64, 90–98. [Google Scholar] [CrossRef]
- Cohen, P.T.; Brewis, N.D.; Hughes, V.; Mann, D.J. Protein serine/threonine phosphatases; an expanding family. FEBS Lett. 1990, 268, 355–359. [Google Scholar] [CrossRef] [Green Version]
- Baskaran, R.; Velmurugan, B.K. Protein phosphatase 2A as therapeutic targets in various disease models. Life Sci. 2018, 210, 40–46. [Google Scholar] [CrossRef]
- Namura, S.; Iihara, K.; Takami, S.; Nagata, I.; Kikuchi, H.; Matsushita, K.; Moskowitz, M.A.; Bonventre, J.V.; Alessandrini, A. Intravenous administration of MEK inhibitor U0126 affords brain protection against forebrain ischemia and focal cerebral ischemia. Proc. Natl. Acad. Sci. USA 2001, 98, 11569–11574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alessandrini, A.; Namura, S.; Moskowitz, M.A.; Bonventre, J.V. MEK1 protein kinase inhibition protects against damage resulting from focal cerebral ischemia. Proc. Natl. Acad. Sci. USA 1999, 96, 12866–12869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Szobi, A.; Lichy, M.; Carnicka, S.; Pancza, D.; Svec, P.; Ravingerova, T.; Adameova, A. Pleiotropic Effects of Simvastatin on Some Calcium Regulatory and Myofibrillar Proteins in Ischemic/Reperfused Heart: Causality of Statins Cardioprotection? Curr. Pharm. Des. 2016, 22, 6451–6458. [Google Scholar] [CrossRef]
- Kronenbitter, A.; Funk, F.; Hackert, K.; Gorreßen, S.; Glaser, D.; Boknik, P.; Poschmann, G.; Stühler, K.; Isić, M.; Krüger, M.; et al. Impaired Ca(2+) cycling of nonischemic myocytes contributes to sarcomere dysfunction early after myocardial infarction. J. Mol. Cell. Cardiol. 2018, 119, 28–39. [Google Scholar] [CrossRef] [PubMed]
- Tian, H.P.; Qiu, T.Z.; Zhao, J.; Li, L.X.; Guo, J. Sphingomyelinase-induced ceramide production stimulate calcium-independent JNK and PP2A activation following cerebral ischemia. Brain Inj. 2009, 23, 1073–1080. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Wu, X.; Xu, J.; Zhou, J.; Han, X.; Guo, J. Src kinase up-regulates the ERK cascade through inactivation of protein phosphatase 2A following cerebral ischemia. BMC Neurosci. 2009, 10, 74. [Google Scholar] [CrossRef] [Green Version]
- Fan, W.J.; van Vuuren, D.; Genade, S.; Lochner, A. Kinases and phosphatases in ischaemic preconditioning: A re-evaluation. Basic Res. Cardiol. 2010, 105, 495–511. [Google Scholar] [CrossRef] [PubMed]
- Martín de la Vega, C.; Burda, J.; Toledo Lobo, M.V.; Salinas, M. Cerebral postischemic reperfusion-induced demethylation of the protein phosphatase 2A catalytic subunit. J. Neurosci. Res 2002, 69, 540–549. [Google Scholar] [CrossRef] [PubMed]
- Penna, C.; Tullio, F.; Perrelli, M.G.; Moro, F.; Abbadessa, G.; Piccione, F.; Carriero, V.; Racca, S.; Pagliaro, P. Ischemia/reperfusion injury is increased and cardioprotection by a postconditioning protocol is lost as cardiac hypertrophy develops in nandrolone treated rats. Basic Res. Cardiol. 2011, 106, 409–420. [Google Scholar] [CrossRef] [Green Version]
- Xu, Z.; Kim, S.; Huh, J. Zinc plays a critical role in the cardioprotective effect of postconditioning by enhancing the activation of the RISK pathway in rat hearts. J. Mol. Cell. Cardiol. 2014, 66, 12–17. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; Qi, Y.; Chen, T. Long-term pre-treatment of antioxidant Ginkgo biloba extract EGb-761 attenuates cerebral-ischemia-induced neuronal damage in aged mice. Biomed. Pharm. 2017, 85, 256–263. [Google Scholar] [CrossRef]
- Zhu, M.; Wang, J.; Liu, M.; Du, D.; Xia, C.; Shen, L.; Zhu, D. Upregulation of protein phosphatase 2A and NR3A-pleiotropic effect of simvastatin on ischemic stroke rats. PLoS ONE 2012, 7, e51552. [Google Scholar] [CrossRef] [Green Version]
- Luo, D.; Fan, X.; Ma, C.; Fan, T.; Wang, X.; Chang, N.; Li, L.; Zhang, Y.; Meng, Z.; Wang, S.; et al. A Study on the Effect of Neurogenesis and Regulation of GSK3β/PP2A Expression in Acupuncture Treatment of Neural Functional Damage Caused by Focal Ischemia in MCAO Rats. Evid. Based Complement. Altern. Med. 2014, 2014, 962343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weinbrenner, C.; Baines, C.P.; Liu, G.S.; Armstrong, S.C.; Ganote, C.E.; Walsh, A.H.; Honkanen, R.E.; Cohen, M.V.; Downey, J.M. Fostriecin, an inhibitor of protein phosphatase 2A, limits myocardial infarct size even when administered after onset of ischemia. Circulation 1998, 98, 899–905. [Google Scholar] [CrossRef] [Green Version]
- Pitt, A.S.; Buchanan, S.K. A Biochemical and Structural Understanding of TOM Complex Interactions and Implications for Human Health and Disease. Cells 2021, 10, 1164. [Google Scholar] [CrossRef]
- Orsini, F.; Moroni, M.; Contursi, C.; Yano, M.; Pelicci, P.; Giorgio, M.; Migliaccio, E. Regulatory effects of the mitochondrial energetic status on mitochondrial p66Shc. Biol. Chem. 2006, 387, 1405–1410. [Google Scholar] [CrossRef]
- Wang, W.; Chen, X.; Zhang, L.; Yi, J.; Ma, Q.; Yin, J.; Zhuo, W.; Gu, J.; Yang, M. Atomic structure of human TOM core complex. Cell Discov. 2020, 6, 67. [Google Scholar] [CrossRef]
- Große, L.; Wurm, C.A.; Brüser, C.; Neumann, D.; Jans, D.C.; Jakobs, S. Bax assembles into large ring-like structures remodeling the mitochondrial outer membrane in apoptosis. Embo J. 2016, 35, 402–413. [Google Scholar] [CrossRef]
- Cartron, P.F.; Bellot, G.; Oliver, L.; Grandier-Vazeille, X.; Manon, S.; Vallette, F.M. Bax inserts into the mitochondrial outer membrane by different mechanisms. FEBS Lett. 2008, 582, 3045–3051. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Ding, Y.; Ye, N.; Wild, C.; Chen, H.; Zhou, J. Direct Activation of Bax Protein for Cancer Therapy. Med. Res. Rev. 2016, 36, 313–341. [Google Scholar] [CrossRef] [PubMed]
- Geisler, S.; Holmström, K.M.; Treis, A.; Skujat, D.; Weber, S.S.; Fiesel, F.C.; Kahle, P.J.; Springer, W. The PINK1/Parkin-mediated mitophagy is compromised by PD-associated mutations. Autophagy 2010, 6, 871–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiesel, F.C.; Caulfield, T.R.; Moussaud-Lamodière, E.L.; Ogaki, K.; Dourado, D.F.; Flores, S.C.; Ross, O.A.; Springer, W. Structural and Functional Impact of Parkinson Disease-Associated Mutations in the E3 Ubiquitin Ligase Parkin. Hum. Mutat. 2015, 36, 774–786. [Google Scholar] [CrossRef] [Green Version]
- Takano, T.; Kohara, M.; Kasama, Y.; Nishimura, T.; Saito, M.; Kai, C.; Tsukiyama-Kohara, K. Translocase of outer mitochondrial membrane 70 expression is induced by hepatitis C virus and is related to the apoptotic response. J. Med. Virol. 2011, 83, 801–809. [Google Scholar] [CrossRef] [PubMed]
- Lazarou, M.; Sliter, D.A.; Kane, L.A.; Sarraf, S.A.; Wang, C.; Burman, J.L.; Sideris, D.P.; Fogel, A.I.; Youle, R.J. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature 2015, 524, 309–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Maio, R.; Barrett, P.J.; Hoffman, E.K.; Barrett, C.W.; Zharikov, A.; Borah, A.; Hu, X.; McCoy, J.; Chu, C.T.; Burton, E.A.; et al. α-Synuclein binds to TOM20 and inhibits mitochondrial protein import in Parkinson’s disease. Sci. Transl. Med. 2016, 8, 342ra378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skulachev, V.P. Cytochrome c in the apoptotic and antioxidant cascades. FEBS Lett. 1998, 423, 275–280. [Google Scholar] [CrossRef] [Green Version]
- Daum, G. Lipids of mitochondria. Biochim. Biophys. Acta 1985, 822, 1–42. [Google Scholar] [CrossRef]
- Gasanoff, E.S.; Yaguzhinsky, L.S.; Garab, G. Cardiolipin, Non-Bilayer Structures and Mitochondrial Bioenergetics: Relevance to Cardiovascular Disease. Cells 2021, 10, 1721. [Google Scholar] [CrossRef]
- Hüttemann, M.; Pecina, P.; Rainbolt, M.; Sanderson, T.H.; Kagan, V.E.; Samavati, L.; Doan, J.W.; Lee, I. The multiple functions of cytochrome c and their regulation in life and death decisions of the mammalian cell: From respiration to apoptosis. Mitochondrion 2011, 11, 369–381. [Google Scholar] [CrossRef] [Green Version]
- Tyurin, V.A.; Tyurina, Y.Y.; Osipov, A.N.; Belikova, N.A.; Basova, L.V.; Kapralov, A.A.; Bayir, H.; Kagan, V.E. Interactions of cardiolipin and lyso-cardiolipins with cytochrome c and tBid: Conflict or assistance in apoptosis. Cell Death Differ. 2007, 14, 872–875. [Google Scholar] [CrossRef] [Green Version]
- Díaz-Quintana, A.; Pérez-Mejías, G.; Guerra-Castellano, A.; De la Rosa, M.A.; Díaz-Moreno, I. Wheel and Deal in the Mitochondrial Inner Membranes: The Tale of Cytochrome c and Cardiolipin. Oxid. Med. Cell. Longev. 2020, 2020, 6813405. [Google Scholar] [CrossRef] [Green Version]
- Kagan, V.E.; Tyurin, V.A.; Jiang, J.; Tyurina, Y.Y.; Ritov, V.B.; Amoscato, A.A.; Osipov, A.N.; Belikova, N.A.; Kapralov, A.A.; Kini, V.; et al. Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors. Nat. Chem. Biol. 2005, 1, 223–232. [Google Scholar] [CrossRef]
- Tyurina, Y.Y.; Poloyac, S.M.; Tyurin, V.A.; Kapralov, A.A.; Jiang, J.; Anthonymuthu, T.S.; Kapralova, V.I.; Vikulina, A.S.; Jung, M.Y.; Epperly, M.W.; et al. A mitochondrial pathway for biosynthesis of lipid mediators. Nat. Chem. 2014, 6, 542–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petrosillo, G.; Ruggiero, F.M.; Paradies, G. Role of reactive oxygen species and cardiolipin in the release of cytochrome c from mitochondria. Faseb J. 2003, 17, 2202–2208. [Google Scholar] [CrossRef] [Green Version]
- Shidoji, Y.; Hayashi, K.; Komura, S.; Ohishi, N.; Yagi, K. Loss of molecular interaction between cytochrome c and cardiolipin due to lipid peroxidation. Biochem. Biophys. Res. Commun. 1999, 264, 343–347. [Google Scholar] [CrossRef]
- Li, P.; Nijhawan, D.; Budihardjo, I.; Srinivasula, S.M.; Ahmad, M.; Alnemri, E.S.; Wang, X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 1997, 91, 479–489. [Google Scholar] [CrossRef] [Green Version]
- Zou, H.; Henzel, W.J.; Liu, X.; Lutschg, A.; Wang, X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 1997, 90, 405–413. [Google Scholar] [CrossRef] [Green Version]
- Green, D.R.; Kroemer, G. The pathophysiology of mitochondrial cell death. Science 2004, 305, 626–629. [Google Scholar] [CrossRef]
- Attardi, L.D.; Reczek, E.E.; Cosmas, C.; Demicco, E.G.; McCurrach, M.E.; Lowe, S.W.; Jacks, T. PERP, an apoptosis-associated target of p53, is a novel member of the PMP-22/gas3 family. Genes Dev. 2000, 14, 704–718. [Google Scholar] [CrossRef]
- Le, S.; Connors, T.J.; Maroney, A.C. c-Jun N-terminal kinase specifically phosphorylates p66ShcA at serine 36 in response to ultraviolet irradiation. J. Biol. Chem. 2001, 276, 48332–48336. [Google Scholar] [CrossRef] [Green Version]
- Natalicchio, A.; De Stefano, F.; Perrini, S.; Laviola, L.; Cignarelli, A.; Caccioppoli, C.; Quagliara, A.; Melchiorre, M.; Leonardini, A.; Conserva, A.; et al. Involvement of the p66Shc protein in glucose transport regulation in skeletal muscle myoblasts. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E228–E237. [Google Scholar] [CrossRef] [Green Version]
- Natalicchio, A.; Tortosa, F.; Perrini, S.; Laviola, L.; Giorgino, F. p66Shc, a multifaceted protein linking Erk signalling, glucose metabolism, and oxidative stress. Arch. Physiol. Biochem. 2011, 117, 116–124. [Google Scholar] [CrossRef]
- Oda, E.; Ohki, R.; Murasawa, H.; Nemoto, J.; Shibue, T.; Yamashita, T.; Tokino, T.; Taniguchi, T.; Tanaka, N. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 2000, 288, 1053–1058. [Google Scholar] [CrossRef] [PubMed]
- Pronsato, L.; Milanesi, L. Effect of testosterone on the regulation of p53 and p66Shc during oxidative stress damage in C2C12 cells. Steroids 2016, 106, 41–54. [Google Scholar] [CrossRef]
- Xu, Y.; Li, N.; Xiang, R.; Sun, P. Emerging roles of the p38 MAPK and PI3K/AKT/mTOR pathways in oncogene-induced senescence. Trends Biochem. Sci. 2014, 39, 268–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Z.J.; Broz, D.K.; Noderer, W.L.; Ferreira, J.P.; Overton, K.W.; Spencer, S.L.; Meyer, T.; Tapscott, S.J.; Attardi, L.D.; Wang, C.L. p53 suppresses muscle differentiation at the myogenin step in response to genotoxic stress. Cell Death Differ. 2015, 22, 560–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haga, S.; Terui, K.; Fukai, M.; Oikawa, Y.; Irani, K.; Furukawa, H.; Todo, S.; Ozaki, M. Preventing hypoxia/reoxygenation damage to hepatocytes by p66(shc) ablation: Up-regulation of anti-oxidant and anti-apoptotic proteins. J. Hepatol. 2008, 48, 422–432. [Google Scholar] [CrossRef]
- Koch, O.R.; Fusco, S.; Ranieri, S.C.; Maulucci, G.; Palozza, P.; Larocca, L.M.; Cravero, A.A.; Farre, S.M.; De Spirito, M.; Galeotti, T.; et al. Role of the life span determinant P66(shcA) in ethanol-induced liver damage. Lab. Investig. 2008, 88, 750–760. [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]
- Smith, W.W.; Norton, D.D.; Gorospe, M.; Jiang, H.; Nemoto, S.; Holbrook, N.J.; Finkel, T.; Kusiak, J.W. Phosphorylation of p66Shc and forkhead proteins mediates Abeta toxicity. J. Cell Biol. 2005, 169, 331–339. [Google Scholar] [CrossRef] [Green Version]
- Wu, Z.; Rogers, B.; Kachi, S.; Hackett, S.F.; Sick, A.; Campochiaro, P.A. Reduction of p66Shc suppresses oxidative damage in retinal pigmented epithelial cells and retina. J. Cell. Physiol. 2006, 209, 996–1005. [Google Scholar] [CrossRef]
- Chahdi, A.; Sorokin, A. Endothelin-1 couples betaPix to p66Shc: Role of betaPix in cell proliferation through FOXO3a phosphorylation and p27kip1 down-regulation independently of Akt. Mol. Biol. Cell. 2008, 19, 2609–2619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camici, G.G.; Schiavoni, M.; Francia, P.; Bachschmid, M.; Martin-Padura, I.; Hersberger, M.; Tanner, F.C.; Pelicci, P.; Volpe, M.; Anversa, P.; et al. Genetic deletion of p66(Shc) adaptor protein prevents hyperglycemia-induced endothelial dysfunction and oxidative stress. Proc. Natl. Acad. Sci. USA 2007, 104, 5217–5222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Francia, P.; delli Gatti, C.; Bachschmid, M.; Martin-Padura, I.; Savoia, C.; Migliaccio, E.; Pelicci, P.G.; Schiavoni, M.; Lüscher, T.F.; Volpe, M.; et al. Deletion of p66shc gene protects against age-related endothelial dysfunction. Circulation 2004, 110, 2889–2895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spescha, R.D.; Glanzmann, M.; Simic, B.; Witassek, F.; Keller, S.; Akhmedov, A.; Tanner, F.C.; Luscher, T.F.; Camici, G.G. Adaptor protein p66(Shc) mediates hypertension-associated, cyclic stretch-dependent, endothelial damage. Hypertension 2014, 64, 347–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vashistha, H.; Singhal, P.C.; Malhotra, A.; Husain, M.; Mathieson, P.; Saleem, M.A.; Kuriakose, C.; Seshan, S.; Wilk, A.; Delvalle, L.; et al. Null mutations at the p66 and bradykinin 2 receptor loci induce divergent phenotypes in the diabetic kidney. Am. J. Physiol. Ren. Physiol. 2012, 303, F1629–F1640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, M.; Mileykovskaya, E.; Dowhan, W. Gluing the respiratory chain together. Cardiolipin is required for supercomplex formation in the inner mitochondrial membrane. J. Biol. Chem. 2002, 277, 43553–43556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raja, V.; Greenberg, M.L. The functions of cardiolipin in cellular metabolism-potential modifiers of the Barth syndrome phenotype. Chem. Phys. Lipids 2014, 179, 49–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dudek, J. Role of Cardiolipin in Mitochondrial Signaling Pathways. Front. Cell Dev. Biol. 2017, 5, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esposti, M.D.; Ngo, A.; Ghelli, A.; Benelli, B.; Carelli, V.; McLennan, H.; Linnane, A.W. The interaction of Q analogs, particularly hydroxydecyl benzoquinone (idebenone), with the respiratory complexes of heart mitochondria. Arch. Biochem. Biophys. 1996, 330, 395–400. [Google Scholar] [CrossRef]
- Fato, R.; Bergamini, C.; Bortolus, M.; Maniero, A.L.; Leoni, S.; Ohnishi, T.; Lenaz, G. Differential effects of mitochondrial Complex I inhibitors on production of reactive oxygen species. Biochim. Biophys. Acta 2009, 1787, 384–392. [Google Scholar] [CrossRef]
- Jaber, S.M.; Ge, S.X.; Milstein, J.L.; VanRyzin, J.W.; Waddell, J.; Polster, B.M. Idebenone Has Distinct Effects on Mitochondrial Respiration in Cortical Astrocytes Compared to Cortical Neurons Due to Differential NQO1 Activity. J. Neurosci. 2020, 40, 4609–4619. [Google Scholar] [CrossRef]
- Sugiyama, Y.; Fujita, T.; Matsumoto, M.; Okamoto, K.; Imada, I. Effects of idebenone (CV-2619) and its metabolites on respiratory activity and lipid peroxidation in brain mitochondria from rats and dogs. J. Pharm. 1985, 8, 1006–1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walter, L.; Nogueira, V.; Leverve, X.; Heitz, M.P.; Bernardi, P.; Fontaine, E. Three classes of ubiquinone analogs regulate the mitochondrial permeability transition pore through a common site. J. Biol. Chem. 2000, 275, 29521–29527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakamura, K.; Murakami, M.; Miura, D.; Yunoki, K.; Enko, K.; Tanaka, M.; Saito, Y.; Nishii, N.; Miyoshi, T.; Yoshida, M.; et al. Beta-Blockers and Oxidative Stress in Patients with Heart Failure. Pharmaceuticals 2011, 4, 1088–1100. [Google Scholar] [CrossRef] [Green Version]
- Tian-Li, Y.; Ruffolo, R.R., Jr.; Feuerstein, G. Antioxidant Action of Carvedilol: A Potential Role in Treatment of Heart Failure. Heart Fail. Rev. 1999, 4, 39–51. [Google Scholar]
- Dandona, P.; Karne, R.; Ghanim, H.; Hamouda, W.; Aljada, A.; Magsino, C.H., Jr. Carvedilol inhibits reactive oxygen species generation by leukocytes and oxidative damage to amino acids. Circulation 2000, 101, 122–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Packer, M.; Bristow, M.R.; Cohn, J.N.; Colucci, W.S.; Fowler, M.B.; Gilbert, E.M.; Shusterman, N.H. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group. N. Engl. J. Med. 1996, 334, 1349–1355. [Google Scholar] [CrossRef] [PubMed]
- Buyse, G.; Mertens, L.; Di Salvo, G.; Matthijs, I.; Weidemann, F.; Eyskens, B.; Goossens, W.; Goemans, N.; Sutherland, G.R.; Van Hove, J.L. Idebenone treatment in Friedreich’s ataxia: Neurological, cardiac, and biochemical monitoring. Neurology 2003, 60, 1679–1681. [Google Scholar] [CrossRef]
- Lyseng-Williamson, K.A. Idebenone: A Review in Leber’s Hereditary Optic Neuropathy. Drugs 2016, 76, 805–813. [Google Scholar] [CrossRef]
- Tomilov, A.; Allen, S.; Hui, C.K.; Bettaieb, A.; Cortopassi, G. Idebenone is a cytoprotective insulin sensitizer whose mechanism is Shc inhibition. Pharm. Res. 2018, 137, 89–103. [Google Scholar] [CrossRef]
- Gestl, E.E.; Anne Bottger, S. Cytoplasmic sequestration of the tumor suppressor p53 by a heat shock protein 70 family member, mortalin, in human colorectal adenocarcinoma cell lines. Biochem. Biophys. Res. Commun. 2012, 423, 411–416. [Google Scholar] [CrossRef]
- Lu, W.J.; Lee, N.P.; Kaul, S.C.; Lan, F.; Poon, R.T.; Wadhwa, R.; Luk, J.M. Mortalin-p53 interaction in cancer cells is stress dependent and constitutes a selective target for cancer therapy. Cell Death Differ. 2011, 18, 1046–1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sadekova, S.; Lehnert, S.; Chow, T.Y. Induction of PBP74/mortalin/Grp75, a member of the hsp70 family, by low doses of ionizing radiation: A possible role in induced radioresistance. Int. J. Radiat. Biol. 1997, 72, 653–660. [Google Scholar] [CrossRef]
- Xu, L.; Voloboueva, L.A.; Ouyang, Y.; Emery, J.F.; Giffard, R.G. Overexpression of mitochondrial Hsp70/Hsp75 in rat brain protects mitochondria, reduces oxidative stress, and protects from focal ischemia. J. Cereb. Blood Flow Metab. 2009, 29, 365–374. [Google Scholar] [CrossRef]
- Ran, Q.; Wadhwa, R.; Kawai, R.; Kaul, S.C.; Sifers, R.N.; Bick, R.J.; Smith, J.R.; Pereira-Smith, O.M. Extramitochondrial localization of mortalin/mthsp70/PBP74/GRP75. Biochem. Biophys. Res. Commun. 2000, 275, 174–179. [Google Scholar] [CrossRef]
- Yang, H.; Zhou, X.; Liu, X.; Yang, L.; Chen, Q.; Zhao, D.; Zuo, J.; Liu, W. Mitochondrial dysfunction induced by knockdown of mortalin is rescued by Parkin. Biochem. Biophys. Res. Commun. 2011, 410, 114–120. [Google Scholar] [CrossRef] [PubMed]
- Havalova, H.; Ondrovicova, G.; Keresztesova, B.; Bauer, J.A.; Pevala, V.; Kutejova, E.; Kunova, N. Mitochondrial HSP70 Chaperone System-The Influence of Post-Translational Modifications and Involvement in Human Diseases. Int. J. Mol. Sci. 2021, 22, 8077. [Google Scholar] [CrossRef]
- Gogia, S.; Neelamegham, S. Role of fluid shear stress in regulating VWF structure, function and related blood disorders. Biorheology 2015, 52, 319–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ganderton, T.; Berndt, M.C.; Chesterman, C.N.; Hogg, P.J. Hypothesis for control of von Willebrand factor multimer size by intra-molecular thiol-disulphide exchange. J. Thromb. Haemost. 2007, 5, 204–206. [Google Scholar] [CrossRef]
- Nagy, P. Kinetics and mechanisms of thiol-disulfide exchange covering direct substitution and thiol oxidation-mediated pathways. Antioxid. Redox Signal. 2013, 18, 1623–1641. [Google Scholar] [CrossRef] [Green Version]
- Giese, B.; Wang, M.; Gao, J.; Stoltz, M.; Müller, P.; Graber, M. Electron relay race in peptides. J. Org. Chem. 2009, 74, 3621–3625. [Google Scholar] [CrossRef]
- Wojtala, A.; Karkucinska-Wieckowska, A.; Sardao, V.A.; Szczepanowska, J.; Kowalski, P.; Pronicki, M.; Duszynski, J.; Wieckowski, M.R. Modulation of mitochondrial dysfunction-related oxidative stress in fibroblasts of patients with Leigh syndrome by inhibition of prooxidative p66Shc pathway. Mitochondrion 2017, 37, 62–79. [Google Scholar] [CrossRef]
- Pang, B.; Zhao, F.; Zhou, Y.; He, B.; Huang, W.; Zhang, F.; Long, Y.G.; Xia, X.; Liu, M.L.; Jiang, Y.H. Systematic Review and Meta-Analysis of the Impact of Hypoxia on Infarcted Myocardium: Better or Worse? Cell. Physiol. Biochem. 2018, 51, 949–960. [Google Scholar] [CrossRef] [PubMed]
- Jain, I.H.; Zazzeron, L.; Goldberger, O.; Marutani, E.; Wojtkiewicz, G.R.; Ast, T.; Wang, H.; Schleifer, G.; Stepanova, A.; Brepoels, K.; et al. Leigh Syndrome Mouse Model Can Be Rescued by Interventions that Normalize Brain Hyperoxia, but Not HIF Activation. Cell Metab. 2019, 30, 824–832.e823. [Google Scholar] [CrossRef]
- Lehrer, S.S.; Fasman, G.D. Ultraviolet irradiation effects in poly-L-tyrosine and model compounds. Identification of bityrosine as a photoproduct. Biochemistry 1967, 6, 757–767. [Google Scholar] [CrossRef] [PubMed]
- Blomberg, M.R. Mechanism of Oxygen Reduction in Cytochrome c Oxidase and the Role of the Active Site Tyrosine. Biochemistry 2016, 55, 489–500. [Google Scholar] [CrossRef]
- Stubbe, J.; van Der Donk, W.A. Protein Radicals in Enzyme Catalysis. Chem. Rev. 1998, 98, 705–762. [Google Scholar] [CrossRef] [PubMed]
- Battistuzzi, G.; Borsari, M.; Cowan, J.A.; Ranieri, A.; Sola, M. Control of cytochrome C redox potential: Axial ligation and protein environment effects. J. Am. Chem. Soc. 2002, 124, 5315–5324. [Google Scholar] [CrossRef] [PubMed]
- Stojanovski, D.; Muller, J.M.; Milenkovic, D.; Guiard, B.; Pfanner, N.; Chacinska, A. The MIA system for protein import into the mitochondrial intermembrane space. Biochim. Biophys. Acta 2008, 1783, 610–617. [Google Scholar] [CrossRef] [Green Version]
- Habich, M.; Salscheider, S.L.; Riemer, J. Cysteine residues in mitochondrial intermembrane space proteins: More than just import. Br. J. Pharm. 2019, 176, 514–531. [Google Scholar] [CrossRef] [Green Version]
- Riemer, J.; Bulleid, N.; Herrmann, J.M. Disulfide formation in the ER and mitochondria: Two solutions to a common process. Science 2009, 324, 1284–1287. [Google Scholar] [CrossRef]
- Trinei, M.; Migliaccio, E.; Bernardi, P.; Paolucci, F.; Pelicci, P.; Giorgio, M. p66Shc, mitochondria, and the generation of reactive oxygen species. Methods Enzym. 2013, 528, 99–110. [Google Scholar] [CrossRef]
- Patriarca, A.; Polticelli, F.; Piro, M.C.; Sinibaldi, F.; Mei, G.; Bari, M.; Santucci, R.; Fiorucci, L. Conversion of cytochrome c into a peroxidase: Inhibitory mechanisms and implication for neurodegenerative diseases. Arch. Biochem. Biophys. 2012, 522, 62–69. [Google Scholar] [CrossRef] [Green Version]
- Baysa, A.; Sagave, J.; Carpi, A.; Zaglia, T.; Campesan, M.; Dahl, C.P.; Bilbija, D.; Troitskaya, M.; Gullestad, L.; Giorgio, M.; et al. The p66ShcA adaptor protein regulates healing after myocardial infarction. Basic Res. Cardiol. 2015, 110, 13. [Google Scholar] [CrossRef] [PubMed]
- Boengler, K.; Bornbaum, J.; Schluter, K.D.; Schulz, R. P66shc and its role in ischemic cardiovascular diseases. Basic Res. Cardiol. 2019, 114, 29. [Google Scholar] [CrossRef]
- Ramraj, S.K.; Elayapillai, S.P.; Pelikan, R.C.; Zhao, Y.D.; Isingizwe, Z.R.; Kennedy, A.L.; Lightfoot, S.A.; Benbrook, D.M. Novel ovarian cancer maintenance therapy targeted at mortalin and mutant p53. Int. J. Cancer 2020, 147, 1086–1097. [Google Scholar] [CrossRef] [PubMed]
- Chandra, V.; Rai, R.; Benbrook, D.M. Utility and Mechanism of SHetA2 and Paclitaxel for Treatment of Endometrial Cancer. Cancers 2021, 13, 2322. [Google Scholar] [CrossRef]
- Benbrook, D.M.; Nammalwar, B.; Long, A.; Matsumoto, H.; Singh, A.; Bunce, R.A.; Berlin, K.D. SHetA2 interference with mortalin binding to p66shc and p53 identified using drug-conjugated magnetic microspheres. Investig. New Drugs 2014, 32, 412–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Zhao, J.; Yang, W.; Bi, Y.; Chi, J.; Tian, J.; Li, W. High-dose alcohol induces reactive oxygen species-mediated apoptosis via PKC-β/p66Shc in mouse primary cardiomyocytes. Biochem. Biophys. Res. Commun. 2015, 456, 656–661. [Google Scholar] [CrossRef]
- Yang, M.; Stowe, D.F.; Udoh, K.B.; Heisner, J.S.; Camara, A.K. Reversible blockade of complex I or inhibition of PKCβ reduces activation and mitochondria translocation of p66Shc to preserve cardiac function after ischemia. PLoS ONE 2014, 9, e113534. [Google Scholar] [CrossRef] [Green Version]
- Virani, S.S.; Alonso, A.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Delling, F.N.; et al. Heart Disease and Stroke Statistics 2014–2020 Update: A Report From the American Heart Association. Circulation 2020, 141, e139–e596. [Google Scholar] [CrossRef]
- Khavjou, O.; Phelps, D.; Leib, A. Projections of Cardiovascular Disease Prevalence and Costs: 2015–2035; RTI International: Research Triangle Park, NC, USA, 2016. [Google Scholar]
- Di Lisa, F.; Menabò, R.; Canton, M.; Petronilli, V. The role of mitochondria in the salvage and the injury of the ischemic myocardium. Biochim. Biophys. Acta 1998, 1366, 69–78. [Google Scholar] [CrossRef] [Green Version]
- Bernardi, P.; Di Lisa, F. The mitochondrial permeability transition pore: Molecular nature and role as a target in cardioprotection. J. Mol. Cell. Cardiol. 2015, 78, 100–106. [Google Scholar] [CrossRef] [PubMed]
- Bolli, R.; Marbán, E. Molecular and cellular mechanisms of myocardial stunning. Physiol. Rev. 1999, 79, 609–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jennings, R.B.; Ganote, C.E.; Reimer, K.A. Ischemic tissue injury. Am J. Pathol. 1975, 81, 179–198. [Google Scholar] [PubMed]
- Ovize, M.; Baxter, G.F.; Di Lisa, F.; Ferdinandy, P.; Garcia-Dorado, D.; Hausenloy, D.J.; Heusch, G.; Vinten-Johansen, J.; Yellon, D.M.; Schulz, R. Postconditioning and protection from reperfusion injury: Where do we stand? Position paper from the Working Group of Cellular Biology of the Heart of the European Society of Cardiology. Cardiovasc. Res. 2010, 87, 406–423. [Google Scholar] [CrossRef] [Green Version]
- Yellon, D.M.; Hausenloy, D.J. Myocardial reperfusion injury. N. Engl. J. Med. 2007, 357, 1121–1135. [Google Scholar] [CrossRef] [PubMed]
- Fadini, G.P.; Albiero, M.; Menegazzo, L.; Boscaro, E.; Pagnin, E.; Iori, E.; Cosma, C.; Lapolla, A.; Pengo, V.; Stendardo, M.; et al. The redox enzyme p66Shc contributes to diabetes and ischemia-induced delay in cutaneous wound healing. Diabetes 2010, 59, 2306–2314. [Google Scholar] [CrossRef] [Green Version]
- Frijhoff, J.; Dagnell, M.; Augsten, M.; Beltrami, E.; Giorgio, M.; Östman, A. The mitochondrial reactive oxygen species regulator p66Shc controls PDGF-induced signaling and migration through protein tyrosine phosphatase oxidation. Free Radic. Biol. Med. 2014, 68, 268–277. [Google Scholar] [CrossRef]
- Obreztchikova, M.; Elouardighi, H.; Ho, M.; Wilson, B.A.; Gertsberg, Z.; Steinberg, S.F. Distinct signaling functions for Shc isoforms in the heart. J. Biol. Chem. 2006, 281, 20197–20204. [Google Scholar] [CrossRef] [Green Version]
- Brown, J.E.; Zeiger, S.L.; Hettinger, J.C.; Brooks, J.D.; Holt, B.; Morrow, J.D.; Musiek, E.S.; Milne, G.; McLaughlin, B. Essential role of the redox-sensitive kinase p66shc in determining energetic and oxidative status and cell fate in neuronal preconditioning. J. Neurosci. 2010, 30, 5242–5252. [Google Scholar] [CrossRef] [Green Version]
- Lalu, M.M.; Csonka, C.; Giricz, Z.; Csont, T.; Schulz, R.; Ferdinandy, P. Preconditioning decreases ischemia/reperfusion-induced release and activation of matrix metalloproteinase-2. Biochem. Biophys. Res. Commun. 2002, 296, 937–941. [Google Scholar] [CrossRef]
- Wang, M.; Zhao, D.; Spinetti, G.; Zhang, J.; Jiang, L.Q.; Pintus, G.; Monticone, R.; Lakatta, E.G. Matrix metalloproteinase 2 activation of transforming growth factor-beta1 (TGF-beta1) and TGF-beta1-type II receptor signaling within the aged arterial wall. Arter. Thromb. Vasc. Biol. 2006, 26, 1503–1509. [Google Scholar] [CrossRef] [Green Version]
- Ahamed, J.; Burg, N.; Yoshinaga, K.; Janczak, C.A.; Rifkin, D.B.; Coller, B.S. In vitro and in vivo evidence for shear-induced activation of latent transforming growth factor-beta1. Blood 2008, 112, 3650–3660. [Google Scholar] [CrossRef] [Green Version]
- Annes, J.P.; Munger, J.S.; Rifkin, D.B. Making sense of latent TGFbeta activation. J. Cell Sci. 2003, 116, 217–224. [Google Scholar] [CrossRef] [Green Version]
- Bujak, M.; Frangogiannis, N.G. The role of TGF-beta signaling in myocardial infarction and cardiac remodeling. Cardiovasc. Res. 2007, 74, 184–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaludercic, N.; Mialet-Perez, J.; Paolocci, N.; Parini, A.; Di Lisa, F. Monoamine oxidases as sources of oxidants in the heart. J. Mol. Cell. Cardiol. 2014, 73, 34–42. [Google Scholar] [CrossRef] [Green Version]
- Kaludercic, N.; Di Lisa, F. Mitochondrial ROS Formation in the Pathogenesis of Diabetic Cardiomyopathy. Front. Cardiovasc. Med. 2020, 7, 12. [Google Scholar] [CrossRef] [Green Version]
- Murphy, M.P.; Smith, R.A. Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu. Rev. Pharm. Toxicol. 2007, 47, 629–656. [Google Scholar] [CrossRef] [PubMed]
- Adlam, V.J.; Harrison, J.C.; Porteous, C.M.; James, A.M.; Smith, R.A.; Murphy, M.P.; Sammut, I.A. Targeting an antioxidant to mitochondria decreases cardiac ischemia-reperfusion injury. Faseb J. 2005, 19, 1088–1095. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.B.; Wander, G.S.; Rastogi, A.; Shukla, P.K.; Mittal, A.; Sharma, J.P.; Mehrotra, S.K.; Kapoor, R.; Chopra, R.K. Randomized, double-blind placebo-controlled trial of coenzyme Q10 in patients with acute myocardial infarction. Cardiovasc. Drugs 1998, 12, 347–353. [Google Scholar] [CrossRef]
- Maulik, N.; Yoshida, T.; Engelman, R.M.; Bagchi, D.; Otani, H.; Das, D.K. Dietary coenzyme Q(10) supplement renders swine hearts resistant to ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2000, 278, H1084–H1090. [Google Scholar] [CrossRef] [PubMed]
- Birnbaum, Y.; Hale, S.L.; Kloner, R.A. The effect of coenzyme Q10 on infarct size in a rabbit model of ischemia/reperfusion. Cardiovasc. Res. 1996, 32, 861–868. [Google Scholar] [CrossRef] [Green Version]
- Whitman, G.J.; Niibori, K.; Yokoyama, H.; Crestanello, J.A.; Lingle, D.M.; Momeni, R. The mechanisms of coenzyme Q10 as therapy for myocardial ischemia reperfusion injury. Mol. Asp. Med. 1997, 18, S195–S203. [Google Scholar] [CrossRef]
- Dikalova, A.E.; Bikineyeva, A.T.; Budzyn, K.; Nazarewicz, R.R.; McCann, L.; Lewis, W.; Harrison, D.G.; Dikalov, S.I. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ. Res. 2010, 107, 106–116. [Google Scholar] [CrossRef] [Green Version]
- Camara, A.K.; Bienengraeber, M.; Stowe, D.F. Mitochondrial approaches to protect against cardiac ischemia and reperfusion injury. Front. Physiol. 2011, 2, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dey, S.; DeMazumder, D.; Sidor, A.; Foster, D.B.; O’Rourke, B. Mitochondrial ROS Drive Sudden Cardiac Death and Chronic Proteome Remodeling in Heart Failure. Circ. Res. 2018, 123, 356–371. [Google Scholar] [CrossRef] [PubMed]
- Ni, R.; Cao, T.; Xiong, S.; Ma, J.; Fan, G.C.; Lacefield, J.C.; Lu, Y.; Le Tissier, S.; Peng, T. Therapeutic inhibition of mitochondrial reactive oxygen species with mito-TEMPO reduces diabetic cardiomyopathy. Free Radic. Biol. Med. 2016, 90, 12–23. [Google Scholar] [CrossRef] [Green Version]
- Birk, A.V.; Chao, W.M.; Bracken, C.; Warren, J.D.; Szeto, H.H. Targeting mitochondrial cardiolipin and the cytochrome c/cardiolipin complex to promote electron transport and optimize mitochondrial ATP synthesis. Br. J. Pharm. 2014, 171, 2017–2028. [Google Scholar] [CrossRef] [Green Version]
- Birk, A.V.; Liu, S.; Soong, Y.; Mills, W.; Singh, P.; Warren, J.D.; Seshan, S.V.; Pardee, J.D.; Szeto, H.H. The mitochondrial-targeted compound SS-31 re-energizes ischemic mitochondria by interacting with cardiolipin. J. Am. Soc. Nephrol. 2013, 24, 1250–1261. [Google Scholar] [CrossRef]
- Kagan, V.E.; Bayir, H.A.; Belikova, N.A.; Kapralov, O.; Tyurina, Y.Y.; Tyurin, V.A.; Jiang, J.; Stoyanovsky, D.A.; Wipf, P.; Kochanek, P.M.; et al. Cytochrome c/cardiolipin relations in mitochondria: A kiss of death. Free Radic. Biol. Med. 2009, 46, 1439–1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kloner, R.A.; Hale, S.L.; Dai, W.; Gorman, R.C.; Shuto, T.; Koomalsingh, K.J.; Gorman, J.H., 3rd; Sloan, R.C.; Frasier, C.R.; Watson, C.A.; et al. Reduction of ischemia/reperfusion injury with bendavia, a mitochondria-targeting cytoprotective Peptide. J. Am. Heart Assoc. 2012, 1, e001644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dai, W.; Shi, J.; Gupta, R.C.; Sabbah, H.N.; Hale, S.L.; Kloner, R.A. Bendavia, a mitochondria-targeting peptide, improves postinfarction cardiac function, prevents adverse left ventricular remodeling, and restores mitochondria-related gene expression in rats. J. Cardiovasc. Pharm. 2014, 64, 543–553. [Google Scholar] [CrossRef]
- Mao, Z.J.; Lin, H.; Hou, J.W.; Zhou, Q.; Wang, Q.; Chen, Y.H. A Meta-Analysis of Resveratrol Protects against Myocardial Ischemia/Reperfusion Injury: Evidence from Small Animal Studies and Insight into Molecular Mechanisms. Oxid. Med. Cell. Longev. 2019, 2019, 5793867. [Google Scholar] [CrossRef]
- Chen, H.Z.; Wan, Y.Z.; Liu, D.P. Cross-talk between SIRT1 and p66Shc in vascular diseases. Trends Cardiovasc. Med. 2013, 23, 237–241. [Google Scholar] [CrossRef] [PubMed]
- Ghatak, A.; Brar, M.J.; Agarwal, A.; Goel, N.; Rastogi, A.K.; Vaish, A.K.; Sircar, A.R.; Chandra, M. Oxy free radical system in heart failure and therapeutic role of oral vitamin E. Int. J. Cardiol. 1996, 57, 119–127. [Google Scholar] [CrossRef]
- McMurray, J.; Chopra, M.; Abdullah, I.; Smith, W.E.; Dargie, H.J. Evidence of oxidative stress in chronic heart failure in humans. Eur. Heart J. 1993, 14, 1493–1498. [Google Scholar] [CrossRef] [PubMed]
- Keith, M.; Geranmayegan, A.; Sole, M.J.; Kurian, R.; Robinson, A.; Omran, A.S.; Jeejeebhoy, K.N. Increased oxidative stress in patients with congestive heart failure. J. Am. Coll. Cardio.l 1998, 31, 1352–1356. [Google Scholar] [CrossRef] [Green Version]
- Belch, J.J.; Bridges, A.B.; Scott, N.; Chopra, M. Oxygen free radicals and congestive heart failure. Br. Heart J. 1991, 65, 245–248. [Google Scholar] [CrossRef] [Green Version]
- Fleisch, J.H.; Spaethe, S.M. Vasodilation and aging evaluated in the isolated perfused rat mesenteric vascular bed: Preliminary observations on the vascular pharmacology of dobutamine. J. Cardiovasc. Pharm. 1981, 3, 187–196. [Google Scholar] [CrossRef]
- Lüscher, T.F.; Tanner, F.C.; Tschudi, M.R.; Noll, G. Endothelial dysfunction in coronary artery disease. Annu. Rev. Med. 1993, 44, 395–418. [Google Scholar] [CrossRef]
- Moritoki, H.; Hosoki, E.; Ishida, Y. Age-related decrease in endothelium-dependent dilator response to histamine in rat mesenteric artery. Eur. J. Pharm. 1986, 126, 61–67. [Google Scholar] [CrossRef]
- Shirasaki, Y.; Su, C.; Lee, T.J.; Kolm, P.; Cline, W.H., Jr.; Nickols, G.A. Endothelial modulation of vascular relaxation to nitrovasodilators in aging and hypertension. J. Pharm. Exp. 1986, 239, 861–866. [Google Scholar]
- Tschudi, M.R.; Barton, M.; Bersinger, N.A.; Moreau, P.; Cosentino, F.; Noll, G.; Malinski, T.; Lüscher, T.F. Effect of age on kinetics of nitric oxide release in rat aorta and pulmonary artery. J. Clin. Investig. 1996, 98, 899–905. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Savarese, G.; Perrone-Filardi, P.; Lüscher, T.F.; Camici, G.G. Enhanced age-dependent cerebrovascular dysfunction is mediated by adaptor protein p66Shc. Int. J. Cardiol. 2014, 175, 446–450. [Google Scholar] [CrossRef]
- Yamamori, T.; White, A.R.; Mattagajasingh, I.; Khanday, F.A.; Haile, A.; Qi, B.; Jeon, B.H.; Bugayenko, A.; Kasuno, K.; Berkowitz, D.E.; et al. P66shc regulates endothelial NO production and endothelium-dependent vasorelaxation: Implications for age-associated vascular dysfunction. J. Mol. Cell. Cardiol. 2005, 39, 992–995. [Google Scholar] [CrossRef]
- 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]
- Cai, W.; He, J.C.; Zhu, L.; Chen, X.; Striker, G.E.; Vlassara, H. AGE-receptor-1 counteracts cellular oxidant stress induced by AGEs via negative regulation of p66shc-dependent FKHRL1 phosphorylation. Am. J. Physiol. Cell Physiol. 2008, 294, C145–C152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menini, S.; Iacobini, C.; Ricci, C.; Oddi, G.; Pesce, C.; Pugliese, F.; Block, K.; Abboud, H.E.; Giorgio, M.; Migliaccio, E.; et al. Ablation of the gene encoding p66Shc protects mice against AGE-induced glomerulopathy by preventing oxidant-dependent tissue injury and further AGE accumulation. Diabetologia 2007, 50, 1997–2007. [Google Scholar] [CrossRef] [Green Version]
- Alikhani, M.; Maclellan, C.M.; Raptis, M.; Vora, S.; Trackman, P.C.; Graves, D.T. Advanced glycation end products induce apoptosis in fibroblasts through activation of ROS, MAP kinases, and the FOXO1 transcription factor. Am. J. Physiol. Cell Physiol. 2007, 292, C850–C856. [Google Scholar] [CrossRef] [PubMed]
- Xi, G.; Shen, X.; Radhakrishnan, Y.; Maile, L.; Clemmons, D. Hyperglycemia-induced p66shc inhibits insulin-like growth factor I-dependent cell survival via impairment of Src kinase-mediated phosphoinositide-3 kinase/AKT activation in vascular smooth muscle cells. Endocrinology 2010, 151, 3611–3623. [Google Scholar] [CrossRef] [Green Version]
- Diaz, M.N.; Frei, B.; Vita, J.A.; Keaney, J.F., Jr. Antioxidants and atherosclerotic heart disease. N. Engl. J. Med. 1997, 337, 408–416. [Google Scholar] [CrossRef]
- Keaney, J.F., Jr.; Shwaery, G.T.; Xu, A.; Nicolosi, R.J.; Loscalzo, J.; Foxall, T.L.; Vita, J.A. 17 beta-estradiol preserves endothelial vasodilator function and limits low-density lipoprotein oxidation in hypercholesterolemic swine. Circulation 1994, 89, 2251–2259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lynch, S.M.; Morrow, J.D.; Roberts, L.J., 2nd; Frei, B. Formation of non-cyclooxygenase-derived prostanoids (F2-isoprostanes) in plasma and low density lipoprotein exposed to oxidative stress in vitro. J. Clin. Investig. 1994, 93, 998–1004. [Google Scholar] [CrossRef] [PubMed]
- Zeiher, A.M.; Schächlinger, V.; Hohnloser, S.H.; Saurbier, B.; Just, H. Coronary atherosclerotic wall thickening and vascular reactivity in humans. Elevated high-density lipoprotein levels ameliorate abnormal vasoconstriction in early atherosclerosis. Circulation 1994, 89, 2525–2532. [Google Scholar] [CrossRef] [Green Version]
- Napoli, C.; Ackah, E.; De Nigris, F.; Del Soldato, P.; D’Armiento, F.P.; Crimi, E.; Condorelli, M.; Sessa, W.C. Chronic treatment with nitric oxide-releasing aspirin reduces plasma low-density lipoprotein oxidation and oxidative stress, arterial oxidation-specific epitopes, and atherogenesis in hypercholesterolemic mice. Proc. Natl. Acad. Sci. USA 2002, 99, 12467–12470. [Google Scholar] [CrossRef] [Green Version]
- Mertens, A.; Verhamme, P.; Bielicki, J.K.; Phillips, M.C.; Quarck, R.; Verreth, W.; Stengel, D.; Ninio, E.; Navab, M.; Mackness, B.; et al. Increased low-density lipoprotein oxidation and impaired high-density lipoprotein antioxidant defense are associated with increased macrophage homing and atherosclerosis in dyslipidemic obese mice: LCAT gene transfer decreases atherosclerosis. Circulation 2003, 107, 1640–1646. [Google Scholar] [CrossRef] [Green Version]
- Ludmer, P.L.; Selwyn, A.P.; Shook, T.L.; Wayne, R.R.; Mudge, G.H.; Alexander, R.W.; Ganz, P. Paradoxical vasoconstriction induced by acetylcholine in atherosclerotic coronary arteries. N. Engl. J. Med. 1986, 315, 1046–1051. [Google Scholar] [CrossRef] [PubMed]
- Miao, Q.; Wang, Q.; Dong, L.; Wang, Y.; Tan, Y.; Zhang, X. The expression of p66shc in peripheral blood monocytes is increased in patients with coronary heart disease and correlated with endothelium-dependent vasodilatation. Heart Vessel. 2015, 30, 451–457. [Google Scholar] [CrossRef] [PubMed]
- Samady, H.; Eshtehardi, P.; McDaniel, M.C.; Suo, J.; Dhawan, S.S.; Maynard, C.; Timmins, L.H.; Quyyumi, A.A.; Giddens, D.P. Coronary artery wall shear stress is associated with progression and transformation of atherosclerotic plaque and arterial remodeling in patients with coronary artery disease. Circulation 2011, 124, 779–788. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Haslem, L.; Hays, J.M.; Hays, F.A. p66Shc in Cardiovascular Pathology. Cells 2022, 11, 1855. https://doi.org/10.3390/cells11111855
Haslem L, Hays JM, Hays FA. p66Shc in Cardiovascular Pathology. Cells. 2022; 11(11):1855. https://doi.org/10.3390/cells11111855
Chicago/Turabian StyleHaslem, Landon, Jennifer M. Hays, and Franklin A. Hays. 2022. "p66Shc in Cardiovascular Pathology" Cells 11, no. 11: 1855. https://doi.org/10.3390/cells11111855
APA StyleHaslem, L., Hays, J. M., & Hays, F. A. (2022). p66Shc in Cardiovascular Pathology. Cells, 11(11), 1855. https://doi.org/10.3390/cells11111855