Mitochondrial Quality Control in Cardiac-Conditioning Strategies against Ischemia-Reperfusion Injury
Abstract
:1. Introduction
2. Protecting the Mitochondria: The Key Target in Myocardial Ischemic Conditioning
3. Mitochondrial Quality Control in Cardioprotection
4. Mitochondrial Biogenesis during Conditioning: It Is Not Too Late to Renew
4.1. Mitochondrial Biogenesis
4.2. Regulation of Mitochondrial Biogenesis by Cardiac Conditioning
5. Fusion and Fission: Two Dynamic Processes in Constant Equilibrium
5.1. Mitochondrial Dynamics
5.1.1. Mitochondrial Fusion
5.1.2. Mitochondrial Fission
5.2. Regulation of Mitochondrial Dynamics by Cardiac Conditioning
6. The Double-Edged Sword of Cardiac Conditioning: “Self-Eating”
6.1. Autophagy
6.2. Regulation of Autophagy by Cardiac Conditioning
7. Mitophagy: The Selective Pathway to Remove Dysfunctional Mitochondria
7.1. Mitophagy
7.2. Regulation of Mitophagy by Cardiac Conditioning
8. Mitochondria-Dependent Cell Death: A Team Work Sacrifice
8.1. Necrosis, Apoptosis, Necroptosis and Pyroptosis Mechanisms
8.1.1. Necrosis
8.1.2. Apoptosis
8.1.3. Necroptosis
8.1.4. Pyroptosis
8.2. Regulation of Mitochondrial-Dependent Cell Death by Cardiac Conditioning
8.2.1. The Main Target of Reperfusion Damage: The mPTP
8.2.2. Inhibiting the Mitochondrial Apoptotic Program
8.2.3. Controlling Necroptosis and Pyroptosis
9. The Role of EVs in Cardiac Conditioning and MQC Mechanisms
9.1. EVs: Microvesicles, Exosomes and Apoptotic Bodies
9.2. Exosomes-Delivery to Protect Heart against Reperfusion Injury
9.3. Pre- and Postconditioning and EVs in Cardiac Protection
9.4. Remote Conditioning and EVs in Cardiac Protection
10. Conclusions and Future Directions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- King, D.R.; Padget, R.L.; Perry, J.; Hoeker, G.; Smyth, J.W.; Brown, D.A.; Poelzing, S. Elevated perfusate [Na+] increases contractile dysfunction during ischemia and reperfusion. Sci. Rep. 2020, 10, 17289. [Google Scholar] [CrossRef]
- Chouchani, E.T.; Pell, V.R.; Gaude, E.; Aksentijevic, D.; Sundier, S.Y.; Robb, E.L.; Logan, A.; Nadtochiy, S.M.; Ord, E.N.J.; Smith, A.C.; et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 2014, 515, 431–435. [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]
- Kurian, G.A.; Rajagopal, R.; Vedantham, S.; Rajesh, M. The role of oxidative stress in myocardial ischemia and reperfusion injury and remodeling: Revisited. Oxid. Med. Cell. Longev. 2016, 2016, 1656450. [Google Scholar] [CrossRef] [Green Version]
- Picca, A.; Mankowski, R.T.; Burman, J.L.; Donisi, L.; Kim, J.S.; Marzetti, E.; Leeuwenburgh, C. Mitochondrial quality control mechanisms as molecular targets in cardiac ageing. Nat. Rev. Cardiol. 2018, 15, 543–554. [Google Scholar] [CrossRef]
- Gil-Hernandez, A.; Silva-Palacios, A. Relevance of endoplasmic reticulum and mitochondria interactions in age-associated diseases. Ageing Res. Rev. 2020, 64, 101193. [Google Scholar] [CrossRef]
- Ferraris, V.A. Commentary: Recovering ischemic myocardium-hibernation, autophagy, preconditioning, mitochondria, stem cells, and more. J. Thorac. Cardiovasc. Surg. 2021, 162, e17–e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, H.; Zhu, P.; Wang, J.; Zhu, H.; Ren, J.; Chen, Y. Pathogenesis of cardiac ischemia reperfusion injury is associated with CK2alpha-disturbed mitochondrial homeostasis via suppression of FUNDC1-related mitophagy. Cell Death Differ. 2018, 25, 1080–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonora, M.; Pinton, P. The mitochondrial permeability transition pore and cancer: Molecular mechanisms involved in cell death. Front. Oncol. 2014, 4, 302. [Google Scholar] [CrossRef] [Green Version]
- Buelna-Chontal, M.; Garcia-Niño, W.R.; Silva-Palacios, A.; Enriquez-Cortina, C.; Zazueta, C. Implications of oxidative and nitrosative post-translational modifications in therapeutic strategies against reperfusion damage. Antioxidants 2021, 10, 749. [Google Scholar] [CrossRef] [PubMed]
- Davidson, S.M.; Riquelme, J.A.; Zheng, Y.; Vicencio, J.M.; Lavandero, S.; Yellon, D.M. Endothelial cells release cardioprotective exosomes that may contribute to ischaemic preconditioning. Sci. Rep. 2018, 8, 15885. [Google Scholar] [CrossRef]
- Yu, P.; Zhang, J.; Yu, S.; Luo, Z.; Hua, F.; Yuan, L.; Zhou, Z.; Liu, Q.; Du, X.; Chen, S.; et al. Protective effect of sevoflurane postconditioning against cardiac ischemia/reperfusion injury via ameliorating mitochondrial impairment, oxidative stress and rescuing autophagic clearance. PLoS ONE 2015, 10, e0134666. [Google Scholar] [CrossRef]
- Zheng, X.; Zu, L.; Becker, L.; Cai, Z.P. Ischemic preconditioning inhibits mitochondrial permeability transition pore opening through the PTEN/PDE4 signaling pathway. Cardiology 2014, 129, 163–173. [Google Scholar] [CrossRef] [Green Version]
- Hausenloy, D.J.; Yellon, D.M. The therapeutic potential of ischemic conditioning: An update. Nat. Rev. Cardiol. 2011, 8, 619–629. [Google Scholar] [CrossRef]
- Roth, S.; Torregroza, C.; Feige, K.; Preckel, B.; Hollmann, M.W.; Weber, N.C.; Huhn, R. Pharmacological conditioning of the heart: An update on experimental developments and clinical implications. Int. J. Mol. Sci. 2021, 22, 2519. [Google Scholar] [CrossRef]
- Chen, G.; Thakkar, M.; Robinson, C.; Dore, S. Limb remote ischemic conditioning: Mechanisms, anesthetics, and the potential for expanding therapeutic options. Front. Neurol. 2018, 9, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mallet, R.T.; Manukhina, E.B.; Ruelas, S.S.; Caffrey, J.L.; Downey, H.F. Cardioprotection by intermittent hypoxia conditioning: Evidence, mechanisms, and therapeutic potential. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H216–H232. [Google Scholar] [CrossRef] [PubMed]
- Zhu, W.Z.; Xie, Y.; Chen, L.; Yang, H.T.; Zhou, Z.N. Intermittent high altitude hypoxia inhibits opening of mitochondrial permeability transition pores against reperfusion injury. J. Mol. Cell. Cardiol. 2006, 40, 96–106. [Google Scholar] [CrossRef]
- Xiao, Y.; Chen, W.; Zhong, Z.; Ding, L.; Bai, H.; Chen, H.; Zhang, H.; Gu, Y.; Lu, S. Electroacupuncture preconditioning attenuates myocardial ischemia-reperfusion injury by inhibiting mitophagy mediated by the mTORC1-ULK1-FUNDC1 pathway. Biomed. Pharmacother. 2020, 127, 110148. [Google Scholar] [CrossRef] [PubMed]
- Yuan, Y.; Pan, S.S. Parkin Mediates Mitophagy to Participate in Cardioprotection Induced by Late Exercise Preconditioning but Bnip3 Does Not. J. Cardiovasc. Pharmacol. 2018, 71, 303–316. [Google Scholar] [CrossRef] [PubMed]
- Hausenloy, D.J.; Yellon, D.M. The second window of preconditioning (SWOP) where are we now? Cardiovasc. Drugs Ther. 2010, 24, 235–254. [Google Scholar] [CrossRef]
- Cohen, M.V.; Yang, X.M.; Downey, J.M. Acidosis, oxygen, and interference with mitochondrial permeability transition pore formation in the early minutes of reperfusion are critical to postconditioning’s success. Basic Res. Cardiol. 2008, 103, 464–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hernandez-Resendiz, S.; Roldan, F.J.; Correa, F.; Martinez-Abundis, E.; Osorio-Valencia, G.; Ruiz-de-Jesus, O.; Alexanderson-Rosas, E.; Vigueras, R.M.; Franco, M.; Zazueta, C. Postconditioning protects against reperfusion injury in hypertensive dilated cardiomyopathy by activating MEK/ERK1/2 signaling. J. Card. Fail. 2013, 19, 135–146. [Google Scholar] [CrossRef] [PubMed]
- Garcia-Niño, W.R.; Correa, F.; Rodriguez-Barrena, J.I.; Leon-Contreras, J.C.; Buelna-Chontal, M.; Soria-Castro, E.; Hernandez-Pando, R.; Pedraza-Chaverri, J.; Zazueta, C. Cardioprotective kinase signaling to subsarcolemmal and interfibrillar mitochondria is mediated by caveolar structures. Basic Res. Cardiol. 2017, 112, 15. [Google Scholar] [CrossRef]
- Hausenloy, D.J.; Maddock, H.L.; Baxter, G.F.; Yellon, D.M. Inhibiting mitochondrial permeability transition pore opening: A new paradigm for myocardial preconditioning? Cardiovasc. Res. 2002, 55, 534–543. [Google Scholar] [CrossRef]
- Hausenloy, D.J.; Yellon, D.M.; Mani-Babu, S.; Duchen, M.R. Preconditioning protects by inhibiting the mitochondrial permeability transition. Am. J. Physiol. Heart Circ. Physiol. 2004, 287, H841–H849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McLeod, C.J.; Jeyabalan, A.P.; Minners, J.O.; Clevenger, R.; Hoyt, R.F., Jr.; Sack, M.N. Delayed ischemic preconditioning activates nuclear-encoded electron-transfer-chain gene expression in parallel with enhanced postanoxic mitochondrial respiratory recovery. Circulation 2004, 110, 534–539. [Google Scholar] [CrossRef] [Green Version]
- Lundberg, K.C.; Szweda, L.I. Preconditioning prevents loss in mitochondrial function and release of cytochrome c during prolonged cardiac ischemia/reperfusion. Arch. Biochem. Biophys 2006, 453, 130–134. [Google Scholar] [CrossRef]
- Velez, D.E.; Hermann, R.; Barreda Frank, M.; Mestre Cordero, V.E.; Savino, E.A.; Varela, A.; Marina Prendes, M.G. Effects of wortmannin on cardioprotection exerted by ischemic preconditioning in rat hearts subjected to ischemia-reperfusion. J. Physiol. Biochem. 2016, 72, 83–91. [Google Scholar] [CrossRef]
- Wu, W.; Zhou, X.; Liu, P.; Fei, W.; Li, L.; Yun, H. Isoflurane reduces hypoxia/reoxygenation-induced apoptosis and mitochondrial permeability transition in rat primary cultured cardiocytes. BMC Anesthesiol. 2014, 14, 17. [Google Scholar] [CrossRef] [Green Version]
- Chang, G.; Zhang, D.; Liu, J.; Zhang, P.; Ye, L.; Lu, K.; Duan, Q.; Zheng, A.; Qin, S. Exenatide protects against hypoxia/reoxygenation-induced apoptosis by improving mitochondrial function in H9c2 cells. Exp. Biol. Med. 2014, 239, 414–422. [Google Scholar] [CrossRef]
- Ljubkovic, M.; Mio, Y.; Marinovic, J.; Stadnicka, A.; Warltier, D.C.; Bosnjak, Z.J.; Bienengraeber, M. Isoflurane preconditioning uncouples mitochondria and protects against hypoxia-reoxygenation. Am. J. Physiol. Cell Physiol. 2007, 292, C1583–C1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Correa, F.; Garcia, N.; Robles, C.; Martinez-Abundis, E.; Zazueta, C. Relationship between oxidative stress and mitochondrial function in the post-conditioned heart. J. Bioenerg. Biomembr. 2008, 40, 599–606. [Google Scholar] [CrossRef] [PubMed]
- Argaud, L.; Gateau-Roesch, O.; Raisky, O.; Loufouat, J.; Robert, D.; Ovize, M. Postconditioning inhibits mitochondrial permeability transition. Circulation 2005, 111, 194–197. [Google Scholar] [CrossRef] [PubMed]
- Jin, C.; Wu, J.; Okada, T. Ischemic postconditioning protects heart against hypoxia-reoxygenation injury via opening of mitochondrial Ca2+-activated K+ channels. J. Mol. Cell. Cardiol. 2008, 45, S30. [Google Scholar] [CrossRef]
- Correa, F.; Buelna-Chontal, M.; Chagoya, V.; Garcia-Rivas, G.; Vigueras, R.M.; Pedraza-Chaverri, J.; Garcia-Nino, W.R.; Hernandez-Pando, R.; Leon-Contreras, J.C.; Zazueta, C. Inhibition of the nitric oxide/cyclic guanosine monophosphate pathway limited the cardioprotective effect of post-conditioning in hearts with apical myocardial infarction. Eur.J. Pharmacol. 2015, 765, 472–481. [Google Scholar] [CrossRef]
- Qiao, S.G.; Sun, Y.; Sun, B.; Wang, A.; Qiu, J.; Hong, L.; An, J.Z.; Wang, C.; Zhang, H.L. Sevoflurane postconditioning protects against myocardial ischemia/reperfusion injury by restoring autophagic flux via an NO-dependent mechanism. Acta Pharmacol. Sin. 2019, 40, 35–45. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.L.; Yao, Y.T.; Fang, N.X.; Zhou, C.H.; Gong, J.S.; Li, L.H. Restoration of autophagic flux in myocardial tissues is required for cardioprotection of sevoflurane postconditioning in rats. Acta Pharmacol. Sin. 2014, 35, 758–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, J.; Xie, H.; Sun, Y.; Zhu, J.; Ying, M.; Qiao, S.; Shao, Q.; Wu, H.; Wang, C. Sevoflurane post-conditioning reduces rat myocardial ischemia reperfusion injury through an increase in NOS and a decrease in phopshorylated NHE1 levels. Int. J. Mol. Med. 2015, 36, 1529–1537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, T.; Yu, S.S.; Zhou, C.Y.; Wang, K.; Wan, Y.C. MicroRNA-206 inhibition and activation of the AMPK/Nampt signalling pathway enhance sevoflurane post-conditioning-induced amelioration of myocardial ischaemia/reperfusion injury. J. Drug Target. 2020, 28, 80–91. [Google Scholar] [CrossRef]
- Yang, L.; Xie, P.; Wu, J.; Yu, J.; Yu, T.; Wang, H.; Wang, J.; Xia, Z.; Zheng, H. Sevoflurane postconditioning improves myocardial mitochondrial respiratory function and reduces myocardial ischemia-reperfusion injury by up-regulating HIF-1. Am. J. Transl. Res. 2016, 8, 4415–4424. [Google Scholar]
- Yu, J.; Wu, J.; Xie, P.; Maimaitili, Y.; Wang, J.; Xia, Z.; Gao, F.; Zhang, X.; Zheng, H. Sevoflurane postconditioning attenuates cardiomyocyte hypoxia/reoxygenation injury via restoring mitochondrial morphology. PeerJ 2016, 4, e2659. [Google Scholar] [CrossRef] [Green Version]
- Shi, X.; Li, Y.; Wang, Y.; Ding, T.; Zhang, X.; Wu, N. Pharmacological postconditioning with sappanone A ameliorates myocardial ischemia reperfusion injury and mitochondrial dysfunction via AMPK-mediated mitochondrial quality control. Toxicol. Appl. Pharmacol. 2021, 427, 115668. [Google Scholar] [CrossRef]
- Yang, Y.; Zhao, L.; Ma, J. Penehyclidine hydrochloride preconditioning provides cardiac protection in a rat model of myocardial ischemia/reperfusion injury via the mechanism of mitochondrial dynamics mechanism. Eur.J. Pharmacol. 2017, 813, 130–139. [Google Scholar] [CrossRef] [PubMed]
- Brodsky, J.L.; Merz, A.; Serio, T. Organelle and proteome quality control mechanisms: How cells are able to keep calm and carry on. Mol. Biol. Cell 2014, 25, 733–734. [Google Scholar] [CrossRef] [Green Version]
- Hurtley, S.; Alderton, G. Quality control in cell biology. Science 2019, 366, 816–817. [Google Scholar] [CrossRef]
- Li, S.; Zhang, J.; Liu, C.; Wang, Q.; Yan, J.; Hui, L.; Jia, Q.; Shan, H.; Tao, L.; Zhang, M. The role of mitophagy in regulating cell death. Oxid. Med. Cell.Longev. 2021, 2021, 6617256. [Google Scholar] [CrossRef]
- Roca-Portoles, A.; Tait, S.W.G. Mitochondrial quality control: From molecule to organelle. Cell Mol. Life Sci. 2021, 78, 3853–3866. [Google Scholar] [CrossRef]
- Sedlackova, L.; Korolchuk, V.I. Mitochondrial quality control as a key determinant of cell survival. Biochim. Biophys Acta Mol. Cell Res. 2019, 1866, 575–587. [Google Scholar] [CrossRef] [PubMed]
- Givvimani, S.; Munjal, C.; Tyagi, N.; Sen, U.; Metreveli, N.; Tyagi, S.C. Mitochondrial division/mitophagy inhibitor (Mdivi) ameliorates pressure overload induced heart failure. PLoS ONE 2012, 7, e32388. [Google Scholar] [CrossRef]
- Yu, W.; Xu, M.; Zhang, T.; Zhang, Q.; Zou, C. Mst1 promotes cardiac ischemia-reperfusion injury by inhibiting the ERK-CREB pathway and repressing FUNDC1-mediated mitophagy. J. Physiol. Sci. 2019, 69, 113–127. [Google Scholar] [CrossRef] [PubMed]
- Ramachandra, C.J.A.; Hernandez-Resendiz, S.; Crespo-Avilan, G.E.; Lin, Y.H.; Hausenloy, D.J. Mitochondria in acute myocardial infarction and cardioprotection. EBioMedicine 2020, 57, 102884. [Google Scholar] [CrossRef] [PubMed]
- Tahrir, F.G.; Langford, D.; Amini, S.; Mohseni Ahooyi, T.; Khalili, K. Mitochondrial quality control in cardiac cells: Mechanisms and role in cardiac cell injury and disease. J. Cell Physiol. 2019, 234, 8122–8133. [Google Scholar] [CrossRef] [PubMed]
- Kulek, A.R.; Anzell, A.; Wider, J.M.; Sanderson, T.H.; Przyklenk, K. Mitochondrial quality control: Role in cardiac models of lethal ischemia-reperfusion injury. Cells 2020, 9, 214. [Google Scholar] [CrossRef] [Green Version]
- Popov, L.D. Mitochondrial biogenesis: An update. J. Cell Mol. Med. 2020, 24, 4892–4899. [Google Scholar] [CrossRef] [Green Version]
- Dominy, J.E.; Puigserver, P. Mitochondrial biogenesis through activation of nuclear signaling proteins. Cold Spring Harb. Perspect. Biol. 2013, 5, a015008. [Google Scholar] [CrossRef]
- Bousette, N.; Kislinger, T.; Fong, V.; Isserlin, R.; Hewel, J.A.; Emil, A.; Gramolini, A.O. Large-scale characterization and analysis of the murine cardiac proteome. J. Proteome Res. 2009, 8, 1887–1901. [Google Scholar] [CrossRef]
- Schmidt, O.; Pfanner, N.; Meisinger, C. Mitochondrial protein import: From proteomics to functional mechanisms. Nat. Rev. Mol. Cell Biol. 2010, 11, 655–667. [Google Scholar] [CrossRef]
- Chew, K.; Zhao, L. Interactions of mitochondrial transcription factor A with DNA damage: Mechanistic insights and functional implications. Genes 2021, 12, 1246. [Google Scholar] [CrossRef] [PubMed]
- Ventura-Clapier, R.; Garnier, A.; Veksler, V. Transcriptional control of mitochondrial biogenesis: The central role of PGC-1alpha. Cardiovasc. Res. 2008, 79, 208–217. [Google Scholar] [CrossRef] [Green Version]
- Fernandez-Marcos, P.J.; Auwerx, J. Regulation of PGC-1alpha, a nodal regulator of mitochondrial biogenesis. Am. J. Clin. Nutr 2011, 93, 884S–890S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gleyzer, N.; Vercauteren, K.; Scarpulla, R.C. Control of mitochondrial transcription specificity factors (TFB1M and TFB2M) by nuclear respiratory factors (NRF-1 and NRF-2) and PGC-1 family coactivators. Mol. Cell Biol. 2005, 25, 1354–1366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schreiber, S.N.; Emter, R.; Hock, M.B.; Knutti, D.; Cardenas, J.; Podvinec, M.; Oakeley, E.J.; Kralli, A. The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc. Natl. Acad. Sci. USA 2004, 101, 6472–6477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, D.; Wang, Y.; Zou, X.; Shi, Y.; Liu, Q.; Huyan, T.; Su, J.; Wang, Q.; Zhang, F.; Li, X.; et al. FOXO1 inhibition prevents renal ischemia-reperfusion injury via cAMP-response element binding protein/PPAR-gamma coactivator-1alpha-mediated mitochondrial biogenesis. Br. J. Pharmacol. 2020, 177, 432–448. [Google Scholar] [CrossRef] [PubMed]
- Yeligar, S.M.; Kang, B.Y.; Bijli, K.M.; Kleinhenz, J.M.; Murphy, T.C.; Torres, G.; San Martin, A.; Sutliff, R.L.; Hart, C.M. PPARgamma regulates mitochondrial structure and function and human pulmonary artery smooth muscle cell proliferation. Am. J. Respir. Cell Mol. Biol. 2018, 58, 648–657. [Google Scholar] [CrossRef] [PubMed]
- Ramachandran, B.; Yu, G.; Gulick, T. Nuclear respiratory factor 1 controls myocyte enhancer factor 2A transcription to provide a mechanism for coordinate expression of respiratory chain subunits. J. Biol. Chem. 2008, 283, 11935–11946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campbell, C.T.; Kolesar, J.E.; Kaufman, B.A. Mitochondrial transcription factor A regulates mitochondrial transcription initiation, DNA packaging, and genome copy number. Biochim. Biophys. Acta 2012, 1819, 921–929. [Google Scholar] [CrossRef] [PubMed]
- Inoue, T.; Ikeda, M.; Ide, T.; Fujino, T.; Matsuo, Y.; Arai, S.; Saku, K.; Sunagawa, K. Twinkle overexpression prevents cardiac rupture after myocardial infarction by alleviating impaired mitochondrial biogenesis. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H509–H519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pisano, A.; Cerbelli, B.; Perli, E.; Pelullo, M.; Bargelli, V.; Preziuso, C.; Mancini, M.; He, L.; Bates, M.G.; Lucena, J.R.; et al. Impaired mitochondrial biogenesis is a common feature to myocardial hypertrophy and end-stage ischemic heart failure. Cardiovasc. Pathol. 2016, 25, 103–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sebastiani, M.; Giordano, C.; Nediani, C.; Travaglini, C.; Borchi, E.; Zani, M.; Feccia, M.; Mancini, M.; Petrozza, V.; Cossarizza, A.; et al. Induction of mitochondrial biogenesis is a maladaptive mechanism in mitochondrial cardiomyopathies. J. Am. Coll. Cardiol. 2007, 50, 1362–1369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karamanlidis, G.; Nascimben, L.; Couper, G.S.; Shekar, P.S.; del Monte, F.; Tian, R. Defective DNA replication impairs mitochondrial biogenesis in human failing hearts. Circ. Res. 2010, 106, 1541–1548. [Google Scholar] [CrossRef] [Green Version]
- Ikeuchi, M.; Matsusaka, H.; Kang, D.; Matsushima, S.; Ide, T.; Kubota, T.; Fujiwara, T.; Hamasaki, N.; Takeshita, A.; Sunagawa, K.; et al. Overexpression of mitochondrial transcription factor a ameliorates mitochondrial deficiencies and cardiac failure after myocardial infarction. Circulation 2005, 112, 683–690. [Google Scholar] [CrossRef] [Green Version]
- Chi, L.; Wang, N.; Yang, W.; Wang, Q.; Zhao, D.; Sun, T.; Li, W. Protection of myocardial ischemia-reperfusion by therapeutic hypercapnia: A mechanism involving improvements in mitochondrial biogenesis and function. J. Cardiovasc. Transl. Res. 2019, 12, 467–477. [Google Scholar] [CrossRef]
- Thomas, A.; Pourpirali, S.; Andres, A.; Mentzer, R.; Gottlieb, R. Mitochondrial biogenesis is driven by protein translation during ischemia and reperfusion. Arch. Cardiovasc. Dis. Suppl. 2017, 9, 160. [Google Scholar] [CrossRef]
- Qi, X.; Wang, J. Melatonin improves mitochondrial biogenesis through the AMPK/PGC1alpha pathway to attenuate ischemia/reperfusion-induced myocardial damage. Aging 2020, 12, 7299–7312. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Liu, C.; Li, J.; Zhou, P.; Zhao, X.; Chen, R.; Song, L.; Zhao, H.; Yan, H. LATS2 deletion attenuates myocardial ischemia-reperfusion injury by promoting mitochondrial biogenesis. Oxid. Med. Cell.Longev. 2021, 2021, 1058872. [Google Scholar] [CrossRef] [PubMed]
- Tan, Y.; Mui, D.; Toan, S.; Zhu, P.; Li, R.; Zhou, H. SERCA overexpression improves mitochondrial quality control and attenuates cardiac microvascular ischemia-reperfusion injury. Mol. Ther. Nucleic Acids 2020, 22, 696–707. [Google Scholar] [CrossRef] [PubMed]
- Basheer, W.A.; Fu, Y.; Shimura, D.; Xiao, S.; Agvanian, S.; Hernandez, D.M.; Hitzeman, T.C.; Hong, T.; Shaw, R.M. Stress response protein GJA1-20k promotes mitochondrial biogenesis, metabolic quiescence, and cardioprotection against ischemia/reperfusion injury. JCI Insight 2018, 3, e121900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, Y.L.; Tao, L.; Peng, F.H.; Zheng, N.Z.; Lin, Q.; Cai, S.Y.; Wang, Q. GJA1-20k attenuates Ang II-induced pathological cardiac hypertrophy by regulating gap junction formation and mitochondrial function. Acta Pharmacol. Sin. 2021, 42, 536–549. [Google Scholar] [CrossRef]
- Fu, Y.; Zhang, S.S.; Xiao, S.; Basheer, W.A.; Baum, R.; Epifantseva, I.; Hong, T.; Shaw, R.M. Cx43 isoform GJA1-20k promotes microtubule dependent mitochondrial transport. Front. Physiol. 2017, 8, 905. [Google Scholar] [CrossRef]
- Sanz, M.N.; Farine, E.; Niederberger, P.; Mendez-Carmona, N.; Wyss, R.K.; Arnold, M.; Gulac, P.; Fiedler, G.M.; Gressette, M.; Garnier, A.; et al. Cardioprotective reperfusion strategies differentially affect mitochondria: Studies in an isolated rat heart model of donation after circulatory death (DCD). Am. J. Transplant. 2019, 19, 331–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, W.; Yang, R.; Yang, J.; Yang, J.; Ding, J.; Wu, H.; Zhang, J. Resveratrol pretreatment protects rat hearts from ischemia/reperfusion injury partly via a NALP3 inflammasome pathway. Int. J. Clin. Exp. Pathol. 2015, 8, 8731–8741. [Google Scholar] [PubMed]
- Tan, L.; Yu, J.T.; Guan, H.S. Resveratrol exerts pharmacological preconditioning by activating PGC-1alpha. Med. Hypotheses 2008, 71, 664–667. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Yang, Z.; Shen, R.; Zhong, W.; Zheng, H.; Chen, Z.; Tang, J.; Zhu, J. Resveratrol improves mitochondrial biogenesis function and activates PGC-1alpha pathway in a preclinical model of early brain injury following subarachnoid hemorrhage. Front. Mol. Biosci. 2021, 8, 620683. [Google Scholar] [CrossRef] [PubMed]
- Biala, A.; Tauriainen, E.; Siltanen, A.; Shi, J.; Merasto, S.; Louhelainen, M.; Martonen, E.; Finckenberg, P.; Muller, D.N.; Mervaala, E. Resveratrol induces mitochondrial biogenesis and ameliorates Ang II-induced cardiac remodeling in transgenic rats harboring human renin and angiotensinogen genes. Blood Press 2010, 19, 196–205. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; He, J.; Ismail, M.; Tweeten, S.; Zeng, F.; Gao, L.; Ballinger, S.; Young, M.; Prabhu, S.D.; Rowe, G.C.; et al. HDAC inhibition induces autophagy and mitochondrial biogenesis to maintain mitochondrial homeostasis during cardiac ischemia/reperfusion injury. J. Mol. Cell. Cardiol. 2019, 130, 36–48. [Google Scholar] [CrossRef]
- Sun, L.; Zhao, M.; Yu, X.J.; Wang, H.; He, X.; Liu, J.K.; Zang, W.J. Cardioprotection by acetylcholine: A novel mechanism via mitochondrial biogenesis and function involving the PGC-1alpha pathway. J. Cell Physiol. 2013, 228, 1238–1248. [Google Scholar] [CrossRef]
- Tilokani, L.; Nagashima, S.; Paupe, V.; Prudent, J. Mitochondrial dynamics: Overview of molecular mechanisms. Essays Biochem. 2018, 62, 341–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.J.; McIntyre, R.L.; Janssens, G.E.; Houtkooper, R.H. Mitochondrial fission and fusion: A dynamic role in aging and potential target for age-related disease. Mech. Ageing Dev. 2020, 186, 111212. [Google Scholar] [CrossRef]
- Chan, D.C. Mitochondrial dynamics and its involvement in disease. Annu. Rev. Pathol. 2020, 15, 235–259. [Google Scholar] [CrossRef] [Green Version]
- Giacomello, M.; Pyakurel, A.; Glytsou, C.; Scorrano, L. The cell biology of mitochondrial membrane dynamics. Nat. Rev. Mol. Cell Biol. 2020, 21, 204–224. [Google Scholar] [CrossRef]
- Koshiba, T.; Detmer, S.A.; Kaiser, J.T.; Chen, H.; McCaffery, J.M.; Chan, D.C. Structural basis of mitochondrial tethering by mitofusin complexes. Science 2004, 305, 858–862. [Google Scholar] [CrossRef] [Green Version]
- Yan, L.; Qi, Y.; Huang, X.; Yu, C.; Lan, L.; Guo, X.; Rao, Z.; Hu, J.; Lou, Z. Structural basis for GTP hydrolysis and conformational change of MFN1 in mediating membrane fusion. Nat. Struct. Mol. Biol. 2018, 25, 233–243. [Google Scholar] [CrossRef]
- Ban, T.; Ishihara, T.; Kohno, H.; Saita, S.; Ichimura, A.; Maenaka, K.; Oka, T.; Mihara, K.; Ishihara, N. Molecular basis of selective mitochondrial fusion by heterotypic action between OPA1 and cardiolipin. Nat. Cell Biol. 2017, 19, 856–863. [Google Scholar] [CrossRef]
- Guillery, O.; Malka, F.; Landes, T.; Guillou, E.; Blackstone, C.; Lombes, A.; Belenguer, P.; Arnoult, D.; Rojo, M. Metalloprotease-mediated OPA1 processing is modulated by the mitochondrial membrane potential. Biol. Cell 2008, 100, 315–325. [Google Scholar] [CrossRef]
- Lackner, L.L. Shaping the dynamic mitochondrial network. BMC Biol. 2014, 12, 35. [Google Scholar] [CrossRef] [Green Version]
- Loson, O.C.; Song, Z.; Chen, H.; Chan, D.C. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol. Biol. Cell 2013, 24, 659–667. [Google Scholar] [CrossRef]
- Kalia, R.; Wang, R.Y.; Yusuf, A.; Thomas, P.V.; Agard, D.A.; Shaw, J.M.; Frost, A. Structural basis of mitochondrial receptor binding and constriction by DRP1. Nature 2018, 558, 401–405. [Google Scholar] [CrossRef]
- Korobova, F.; Ramabhadran, V.; Higgs, H.N. An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science 2013, 339, 464–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cho, B.; Cho, H.M.; Jo, Y.; Kim, H.D.; Song, M.; Moon, C.; Kim, H.; Kim, K.; Sesaki, H.; Rhyu, I.J.; et al. Constriction of the mitochondrial inner compartment is a priming event for mitochondrial division. Nat. Commun. 2017, 8, 15754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maneechote, C.; Palee, S.; Kerdphoo, S.; Jaiwongkam, T.; Chattipakorn, S.C.; Chattipakorn, N. Balancing mitochondrial dynamics via increasing mitochondrial fusion attenuates infarct size and left ventricular dysfunction in rats with cardiac ischemia/reperfusion injury. Clin. Sci. 2019, 133, 497–513. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Zhai, M.; Xu, S.; Ponnusamy, M.; Huang, Y.; Liu, C.Y.; Wang, M.; Shan, C.; Shan, P.P.; Gao, X.Q.; et al. NFATc3-dependent expression of miR-153-3p promotes mitochondrial fragmentation in cardiac hypertrophy by impairing mitofusin-1 expression. Theranostics 2020, 10, 553–566. [Google Scholar] [CrossRef] [PubMed]
- Hsiao, Y.T.; Shimizu, I.; Wakasugi, T.; Yoshida, Y.; Ikegami, R.; Hayashi, Y.; Suda, M.; Katsuumi, G.; Nakao, M.; Ozawa, T.; et al. Cardiac mitofusin-1 is reduced in non-responding patients with idiopathic dilated cardiomyopathy. Sci. Rep. 2021, 11, 6722. [Google Scholar] [CrossRef]
- Wu, Q.R.; Zheng, D.L.; Liu, P.M.; Yang, H.; Li, L.A.; Kuang, S.J.; Lai, Y.Y.; Rao, F.; Xue, Y.M.; Lin, J.J.; et al. High glucose induces Drp1-mediated mitochondrial fission via the Orai1 calcium channel to participate in diabetic cardiomyocyte hypertrophy. Cell Death Dis. 2021, 12, 216. [Google Scholar] [CrossRef]
- Sumida, M.; Doi, K.; Ogasawara, E.; Yamashita, T.; Hamasaki, Y.; Kariya, T.; Takimoto, E.; Yahagi, N.; Nangaku, M.; Noiri, E. Regulation of mitochondrial dynamics by dynamin-related protein-1 in acute cardiorenal syndrome. J. Am. Soc. Nephrol. 2015, 26, 2378–2387. [Google Scholar] [CrossRef] [Green Version]
- Cooper, H.A.; Eguchi, S. Inhibition of mitochondrial fission as a novel therapeutic strategy to reduce mortality upon myocardial infarction. Clin. Sci. 2018, 132, 2163–2167. [Google Scholar] [CrossRef]
- Ong, S.B.; Subrayan, S.; Lim, S.Y.; Yellon, D.M.; Davidson, S.M.; Hausenloy, D.J. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation 2010, 121, 2012–2022. [Google Scholar] [CrossRef] [Green Version]
- Bouche, L.; Kamel, R.; Tamareille, S.; Garcia, G.; Villedieu, C.; Pillot, B.; Gueguen, N.; Chehaitly, A.; Chao de la Barca, J.M.; Beaumont, J.; et al. DRP1 haploinsufficiency attenuates cardiac ischemia/reperfusion injuries. PLoS ONE 2021, 16, e0248554. [Google Scholar] [CrossRef]
- Sharp, W.W.; Fang, Y.H.; Han, M.; Zhang, H.J.; Hong, Z.; Banathy, A.; Morrow, E.; Ryan, J.J.; Archer, S.L. Dynamin-related protein 1 (Drp1)-mediated diastolic dysfunction in myocardial ischemia-reperfusion injury: Therapeutic benefits of Drp1 inhibition to reduce mitochondrial fission. FASEB J. 2014, 28, 316–326. [Google Scholar] [CrossRef] [Green Version]
- Papanicolaou, K.N.; Khairallah, R.J.; Ngoh, G.A.; Chikando, A.; Luptak, I.; O’Shea, K.M.; Riley, D.D.; Lugus, J.J.; Colucci, W.S.; Lederer, W.J.; et al. Mitofusin-2 maintains mitochondrial structure and contributes to stress-induced permeability transition in cardiac myocytes. Mol. Cell Biol. 2011, 31, 1309–1328. [Google Scholar] [CrossRef] [Green Version]
- Hall, A.R.; Burke, N.; Dongworth, R.K.; Kalkhoran, S.B.; Dyson, A.; Vicencio, J.M.; Dorn, G.W., II; Yellon, D.M.; Hausenloy, D.J. Hearts deficient in both Mfn1 and Mfn2 are protected against acute myocardial infarction. Cell Death Dis. 2016, 7, e2238. [Google Scholar] [CrossRef]
- Song, M.; Mihara, K.; Chen, Y.; Scorrano, L.; Dorn, G.W., 2nd. Mitochondrial fission and fusion factors reciprocally orchestrate mitophagic culling in mouse hearts and cultured fibroblasts. Cell Metab. 2015, 21, 273–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Civiletto, G.; Varanita, T.; Cerutti, R.; Gorletta, T.; Barbaro, S.; Marchet, S.; Lamperti, C.; Viscomi, C.; Scorrano, L.; Zeviani, M. Opa1 overexpression ameliorates the phenotype of two mitochondrial disease mouse models. Cell Metab. 2015, 21, 845–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Acin-Perez, R.; Lechuga-Vieco, A.V.; Del Mar Munoz, M.; Nieto-Arellano, R.; Torroja, C.; Sanchez-Cabo, F.; Jimenez, C.; Gonzalez-Guerra, A.; Carrascoso, I.; Beninca, C.; et al. Ablation of the stress protease OMA1 protects against heart failure in mice. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Dorn, G.W., 2nd. PINK1-phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria. Science 2013, 340, 471–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McLelland, G.L.; Goiran, T.; Yi, W.; Dorval, G.; Chen, C.X.; Lauinger, N.D.; Krahn, A.I.; Valimehr, S.; Rakovic, A.; Rouiller, I.; et al. Mfn2 ubiquitination by PINK1/parkin gates the p97-dependent release of ER from mitochondria to drive mitophagy. Elife 2018, 7, e32866. [Google Scholar] [CrossRef]
- Samant, S.A.; Zhang, H.J.; Hong, Z.; Pillai, V.B.; Sundaresan, N.R.; Wolfgeher, D.; Archer, S.L.; Chan, D.C.; Gupta, M.P. SIRT3 deacetylates and activates OPA1 to regulate mitochondrial dynamics during stress. Mol. Cell Biol. 2014, 34, 807–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cereghetti, G.M.; Stangherlin, A.; Martins de Brito, O.; Chang, C.R.; Blackstone, C.; Bernardi, P.; Scorrano, L. Dephosphorylation by calcineurin regulates translocation of Drp1 to mitochondria. Proc. Natl. Acad. Sci. USA 2008, 105, 15803–15808. [Google Scholar] [CrossRef] [Green Version]
- Pasdois, P.; Parker, J.E.; Halestrap, A.P. Extent of mitochondrial hexokinase II dissociation during ischemia correlates with mitochondrial cytochrome c release, reactive oxygen species production, and infarct size on reperfusion. J. Am. Heart Assoc. 2012, 2, e005645. [Google Scholar] [CrossRef] [Green Version]
- Pereira, G.C.; Lee, L.; Rawlings, N.; Ouwendijk, J.; Parker, J.E.; Andrienko, T.N.; Henley, J.M.; Halestrap, A.P. Hexokinase II dissociation alone cannot account for changes in heart mitochondrial function, morphology and sensitivity to permeability transition pore opening following ischemia. PLoS ONE 2020, 15, e0234653. [Google Scholar] [CrossRef]
- Cellier, L.; Tamareille, S.; Kalakech, H.; Guillou, S.; Lenaers, G.; Prunier, F.; Mirebeau-Prunier, D. Remote ischemic conditioning influences mitochondrial dynamics. Shock 2016, 45, 192–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le Page, S.; Niro, M.; Fauconnier, J.; Cellier, L.; Tamareille, S.; Gharib, A.; Chevrollier, A.; Loufrani, L.; Grenier, C.; Kamel, R.; et al. Increase in cardiac ischemia-reperfusion injuries in Opa1+/− mouse model. PLoS ONE 2016, 11, e0164066. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.D.; Chen, H.J.; Wang, D.L.; Wang, H.; Deng, Q. Pim-1 kinase regulating dynamics related protein 1 mediates sevoflurane postconditioning-induced cardioprotection. Chin. Med. J. 2017, 130, 309–317. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.; Maimaitili, Y.; Xie, P.; Wu, J.J.; Wang, J.; Yang, Y.N.; Ma, H.P.; Zheng, H. High glucose concentration abrogates sevoflurane post-conditioning cardioprotection by advancing mitochondrial fission but dynamin-related protein 1 inhibitor restores these effects. Acta Physiol. 2017, 220, 83–98. [Google Scholar] [CrossRef] [PubMed]
- Kamga Pride, C.; Mo, L.; Quesnelle, K.; Dagda, R.K.; Murillo, D.; Geary, L.; Corey, C.; Portella, R.; Zharikov, S.; St Croix, C.; et al. Nitrite activates protein kinase A in normoxia to mediate mitochondrial fusion and tolerance to ischaemia/reperfusion. Cardiovasc. Res. 2014, 101, 57–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khuanjing, T.; Palee, S.; Kerdphoo, S.; Jaiwongkam, T.; Anomasiri, A.; Chattipakorn, S.C.; Chattipakorn, N. Donepezil attenuated cardiac ischemia/reperfusion injury through balancing mitochondrial dynamics, mitophagy, and autophagy. Transl. Res. 2021, 230, 82–97. [Google Scholar] [CrossRef] [PubMed]
- Ikeda, Y.; Shirakabe, A.; Maejima, Y.; Zhai, P.; Sciarretta, S.; Toli, J.; Nomura, M.; Mihara, K.; Egashira, K.; Ohishi, M.; et al. Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ. Res. 2015, 116, 264–278. [Google Scholar] [CrossRef] [PubMed]
- Kalkhoran, S.B.; Kriston-Vizi, J.; Hernandez-Resendiz, S.; Crespo-Avilan, G.E.; Rosdah, A.A.; Lees, J.G.; Da Costa, J.R.S.; Ling, N.X.Y.; Holien, J.K.; Samangouei, P.; et al. Hydralazine protects the heart against acute ischemia/reperfusion injury by inhibiting Drp1-mediated mitochondrial fission. Cardiovasc. Res. 2021. [Google Scholar] [CrossRef] [PubMed]
- Dube, K.; Dhanabalan, K.; Salie, R.; Blignaut, M.; Huisamen, B.; Lochner, A. Melatonin has profound effects on mitochondrial dynamics in myocardial ischaemia/reperfusion. Heliyon 2019, 5, e02659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bai, Y.; Yang, Y.; Gao, Y.; Lin, D.; Wang, Z.; Ma, J. Melatonin postconditioning ameliorates anoxia/reoxygenation injury by regulating mitophagy and mitochondrial dynamics in a SIRT3-dependent manner. Eur.J. Pharmacol. 2021, 904, 174157. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Wang, Y.; Xu, J.; Tian, F.; Hu, S.; Chen, Y.; Fu, Z. Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J. Pineal Res. 2019, 66, e12542. [Google Scholar] [CrossRef]
- Chun, Y.; Kim, J. Autophagy: An essential degradation program for cellular homeostasis and life. Cells 2018, 7, 278. [Google Scholar] [CrossRef] [Green Version]
- Karanasios, E.; Walker, S.A.; Okkenhaug, H.; Manifava, M.; Hummel, E.; Zimmermann, H.; Ahmed, Q.; Domart, M.C.; Collinson, L.; Ktistakis, N.T. Autophagy initiation by ULK complex assembly on ER tubulovesicular regions marked by ATG9 vesicles. Nat. Commun. 2016, 7, 12420. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zachari, M.; Ganley, I.G. The mammalian ULK1 complex and autophagy initiation. Essays Biochem. 2017, 61, 585–596. [Google Scholar] [CrossRef] [Green Version]
- Roberts, R.; Ktistakis, N.T. Omegasomes: PI3P platforms that manufacture autophagosomes. Essays Biochem. 2013, 55, 17–27. [Google Scholar] [CrossRef] [PubMed]
- Feng, Y.; He, D.; Yao, Z.; Klionsky, D.J. The machinery of macroautophagy. Cell Res. 2014, 24, 24–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sawa-Makarska, J.; Baumann, V.; Coudevylle, N.; von Bulow, S.; Nogellova, V.; Abert, C.; Schuschnig, M.; Graef, M.; Hummer, G.; Martens, S. Reconstitution of autophagosome nucleation defines Atg9 vesicles as seeds for membrane formation. Science 2020, 369, eaaz7714. [Google Scholar] [CrossRef] [PubMed]
- Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef]
- Wirth, M.; Zhang, W.; Razi, M.; Nyoni, L.; Joshi, D.; O’Reilly, N.; Johansen, T.; Tooze, S.A.; Mouilleron, S. Molecular determinants regulating selective binding of autophagy adapters and receptors to ATG8 proteins. Nat. Commun. 2019, 10, 2055. [Google Scholar] [CrossRef] [Green Version]
- Walczak, M.; Martens, S. Dissecting the role of the Atg12-Atg5-Atg16 complex during autophagosome formation. Autophagy 2013, 9, 424–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mauvezin, C.; Nagy, P.; Juhasz, G.; Neufeld, T.P. Autophagosome-lysosome fusion is independent of V-ATPase-mediated acidification. Nat. Commun. 2015, 6, 7007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salazar, G.; Cullen, A.; Huang, J.; Zhao, Y.; Serino, A.; Hilenski, L.; Patrushev, N.; Forouzandeh, F.; Hwang, H.S. SQSTM1/p62 and PPARGC1A/PGC-1alpha at the interface of autophagy and vascular senescence. Autophagy 2020, 16, 1092–1110. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Liu, C.; Gu, L.; Wang, L.; Shang, Y.; Liu, Q.; Wan, J.; Shi, J.; Wang, F.; Xu, Z.; et al. Autophagy protects cardiomyocytes from the myocardial ischaemia-reperfusion injury through the clearance of CLP36. Open Biol. 2016, 6, 160177. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Liu, H.; Foyil, S.R.; Godar, R.J.; Weinheimer, C.J.; Hill, J.A.; Diwan, A. Impaired autophagosome clearance contributes to cardiomyocyte death in ischemia/reperfusion injury. Circulation 2012, 125, 3170–3181. [Google Scholar] [CrossRef] [Green Version]
- McKnight, N.C.; Zhenyu, Y. Beclin 1, an essential component and master regulator of PI3K-III in health and disease. Curr. Pathobiol. Rep. 2013, 1, 231–238. [Google Scholar] [CrossRef] [Green Version]
- Cui, L.; Zhao, L.P.; Ye, J.Y.; Yang, L.; Huang, Y.; Jiang, X.P.; Zhang, Q.; Jia, J.Z.; Zhang, D.X.; Huang, Y. The lysosomal membrane protein Lamp2 alleviates lysosomal cell death by promoting autophagic flux in ischemic cardiomyocytes. Front. Cell Dev. Biol. 2020, 8, 31. [Google Scholar] [CrossRef] [Green Version]
- Ma, X.; Liu, H.; Foyil, S.R.; Godar, R.J.; Weinheimer, C.J.; Diwan, A. Autophagy is impaired in cardiac ischemia-reperfusion injury. Autophagy 2012, 8, 1394–1396. [Google Scholar] [CrossRef] [Green Version]
- Gidlof, O.; Johnstone, A.L.; Bader, K.; Khomtchouk, B.B.; O’Reilly, J.J.; Celik, S.; Van Booven, D.J.; Wahlestedt, C.; Metzler, B.; Erlinge, D. Ischemic preconditioning confers epigenetic repression of mTOR and induction of autophagy through G9a-dependent H3K9 dimethylation. J. Am. Heart Assoc. 2016, 5, e004076. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Y.; Gu, S.; Li, X.; Tan, J.; Liu, S.; Jiang, Y.; Zhang, C.; Gao, L.; Yang, H.T. Berbamine postconditioning protects the heart from ischemia/reperfusion injury through modulation of autophagy. Cell Death Dis. 2017, 8, e2577. [Google Scholar] [CrossRef] [Green Version]
- Gurusamy, N.; Lekli, I.; Gorbunov, N.V.; Gherghiceanu, M.; Popescu, L.M.; Das, D.K. Cardioprotection by adaptation to ischaemia augments autophagy in association with BAG-1 protein. J. Cell Mol. Med. 2009, 13, 373–387. [Google Scholar] [CrossRef] [Green Version]
- Huang, C.; Yitzhaki, S.; Perry, C.N.; Liu, W.; Giricz, Z.; Mentzer, R.M., Jr.; Gottlieb, R.A. Autophagy induced by ischemic preconditioning is essential for cardioprotection. J. Cardiovasc. Transl. Res. 2010, 3, 365–373. [Google Scholar] [CrossRef] [Green Version]
- Shao, J.; Miao, C.; Geng, Z.; Gu, M.; Wu, Y.; Li, Q. Effect of eNOS on ischemic postconditioning-induced autophagy against ischemia/reperfusion injury in mice. Biomed. Res. Int. 2019, 2019, 5201014. [Google Scholar] [CrossRef]
- Wei, C.; Li, H.; Han, L.; Zhang, L.; Yang, X. Activation of autophagy in ischemic postconditioning contributes to cardioprotective effects against ischemia/reperfusion injury in rat hearts. J. Cardiovasc. Pharmacol. 2013, 61, 416–422. [Google Scholar] [CrossRef]
- Guo, L.; Xu, J.M.; Mo, X.Y. Ischemic postconditioning regulates cardiomyocyte autophagic activity following ischemia/reperfusion injury. Mol. Med. Rep. 2015, 12, 1169–1176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yitzhaki, S.; Huang, C.; Liu, W.; Lee, Y.; Gustafsson, A.B.; Mentzer, R.M., Jr.; Gottlieb, R.A. Autophagy is required for preconditioning by the adenosine A1 receptor-selective agonist CCPA. Basic Res. Cardiol. 2009, 104, 157–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, C.; Liu, W.; Perry, C.N.; Yitzhaki, S.; Lee, Y.; Yuan, H.; Tsukada, Y.T.; Hamacher-Brady, A.; Mentzer, R.M., Jr.; Gottlieb, R.A. Autophagy and protein kinase C are required for cardioprotection by sulfaphenazole. Am. J. Physiol. Heart Circ. Physiol. 2010, 298, H570–H579. [Google Scholar] [CrossRef] [Green Version]
- Andres, A.M.; Hernandez, G.; Lee, P.; Huang, C.; Ratliff, E.P.; Sin, J.; Thornton, C.A.; Damasco, M.V.; Gottlieb, R.A. Mitophagy is required for acute cardioprotection by simvastatin. Antioxid. Redox Signal. 2014, 21, 1960–1973. [Google Scholar] [CrossRef]
- Qiao, S.; Xie, H.; Wang, C.; Wu, X.; Liu, H.; Liu, C. Delayed anesthetic preconditioning protects against myocardial infarction via activation of nuclear factor-kappaB and upregulation of autophagy. J. Anesth. 2013, 27, 251–260. [Google Scholar] [CrossRef]
- Shiomi, M.; Miyamae, M.; Takemura, G.; Kaneda, K.; Inamura, Y.; Onishi, A.; Koshinuma, S.; Momota, Y.; Minami, T.; Figueredo, V.M. Sevoflurane induces cardioprotection through reactive oxygen species-mediated upregulation of autophagy in isolated guinea pig hearts. J. Anesth. 2014, 28, 593–600. [Google Scholar] [CrossRef] [Green Version]
- Hong, L.; Sun, Y.; An, J.Z.; Wang, C.; Qiao, S.G. Sevoflurane preconditioning confers delayed cardioprotection by upregulating AMP-activated protein kinase levels to restore autophagic flux in ischemia-reperfusion rat hearts. Med. Sci. Monit. 2020, 26, e922176. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.; Bu, M.; Yun, H. Sevoflurane prevents hypoxia/reoxygenation-induced cardiomyocyte apoptosis by inhibiting PI3KC3-mediated autophagy. Hum. Cell 2019, 32, 150–159. [Google Scholar] [CrossRef] [PubMed]
- Shi, H.H.; Wang, Z.Q.; Zhang, S. MiR-208a participates with sevoflurane post-conditioning in protecting neonatal rat cardiomyocytes with simulated ischemia-reperfusion injury via PI3K/AKT signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 943–955. [Google Scholar] [CrossRef] [PubMed]
- Choi, G.E.; Lee, H.J.; Chae, C.W.; Cho, J.H.; Jung, Y.H.; Kim, J.S.; Kim, S.Y.; Lim, J.R.; Han, H.J. BNIP3L/NIX-mediated mitophagy protects against glucocorticoid-induced synapse defects. Nat. Commun. 2021, 12, 487. [Google Scholar] [CrossRef] [PubMed]
- Shiba-Fukushima, K.; Imai, Y.; Yoshida, S.; Ishihama, Y.; Kanao, T.; Sato, S.; Hattori, N. PINK1-mediated phosphorylation of the Parkin ubiquitin-like domain primes mitochondrial translocation of Parkin and regulates mitophagy. Sci. Rep. 2012, 2, 1002. [Google Scholar] [CrossRef] [PubMed]
- Kane, L.A.; Lazarou, M.; Fogel, A.I.; Li, Y.; Yamano, K.; Sarraf, S.A.; Banerjee, S.; Youle, R.J. PINK1 phosphorylates ubiquitin to activate Parkin E3 ubiquitin ligase activity. J. Cell Biol. 2014, 205, 143–153. [Google Scholar] [CrossRef] [PubMed]
- Narendra, D.; Kane, L.A.; Hauser, D.N.; Fearnley, I.M.; Youle, R.J. p62/SQSTM1 is required for Parkin-induced mitochondrial clustering but not mitophagy; VDAC1 is dispensable for both. Autophagy 2010, 6, 1090–1106. [Google Scholar] [CrossRef] [PubMed]
- O’Sullivan, T.E.; Johnson, L.R.; Kang, H.H.; Sun, J.C. BNIP3- and BNIP3L-mediated mitophagy promotes the generation of natural killer cell memory. Immunity 2015, 43, 331–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marinkovic, M.; Sprung, M.; Novak, I. Dimerization of mitophagy receptor BNIP3L/NIX is essential for recruitment of autophagic machinery. Autophagy 2021, 17, 1232–1243. [Google Scholar] [CrossRef] [PubMed]
- Hanna, R.A.; Quinsay, M.N.; Orogo, A.M.; Giang, K.; Rikka, S.; Gustafsson, A.B. Microtubule-associated protein 1 light chain 3 (LC3) interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy. J. Biol. Chem. 2012, 287, 19094–19104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, L.; Feng, D.; Chen, G.; Chen, M.; Zheng, Q.; Song, P.; Ma, Q.; Zhu, C.; Wang, R.; Qi, W.; et al. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat. Cell Biol. 2012, 14, 177–185. [Google Scholar] [CrossRef] [PubMed]
- Padman, B.S.; Nguyen, T.N.; Uoselis, L.; Skulsuppaisarn, M.; Nguyen, L.K.; Lazarou, M. LC3/GABARAPs drive ubiquitin-independent recruitment of Optineurin and NDP52 to amplify mitophagy. Nat. Commun. 2019, 10, 408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, Y.; Chiang, W.C.; Sumpter, R., Jr.; Mishra, P.; Levine, B. Prohibitin 2 Is an Inner Mitochondrial Membrane Mitophagy Receptor. Cell 2017, 168, 224–238.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Hernandez-Resendiz, S.; Prunier, F.; Girao, H.; Dorn, G.; Hausenloy, D.J.; Action, E.-C.C. Targeting mitochondrial fusion and fission proteins for cardioprotection. J. Cell Mol. Med. 2020, 24, 6571–6585. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Siraj, S.; Zhang, R.; Chen, Q. Mitophagy receptor FUNDC1 regulates mitochondrial homeostasis and protects the heart from I/R injury. Autophagy 2017, 13, 1080–1081. [Google Scholar] [CrossRef] [PubMed]
- Siddall, H.K.; Yellon, D.M.; Ong, S.B.; Mukherjee, U.A.; Burke, N.; Hall, A.R.; Angelova, P.R.; Ludtmann, M.H.; Deas, E.; Davidson, S.M.; et al. Loss of PINK1 increases the heart’s vulnerability to ischemia-reperfusion injury. PLoS ONE 2013, 8, e62400. [Google Scholar] [CrossRef]
- Kubli, D.A.; Zhang, X.; Lee, Y.; Hanna, R.A.; Quinsay, M.N.; Nguyen, C.K.; Jimenez, R.; Petrosyan, S.; Murphy, A.N.; Gustafsson, A.B. Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction. J. Biol. Chem. 2013, 288, 915–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, T.L.; Lee, M.H.; Chen, Y.C.; Lee, Y.C.; Lai, T.C.; Lin, H.Y.; Hsu, L.F.; Sung, H.C.; Lee, C.W.; Chen, Y.L. Vitamin D attenuates ischemia/reperfusion-induced cardiac injury by reducing mitochondrial fission and mitophagy. Front. Pharmacol. 2020, 11, 604700. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.; Lee, H.Y.; Hanna, R.A.; Gustafsson, A.B. Mitochondrial autophagy by Bnip3 involves Drp1-mediated mitochondrial fission and recruitment of Parkin in cardiac myocytes. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H1924–H1931. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Godar, R.J.; Liu, H.; Diwan, A. Enhancing lysosome biogenesis attenuates BNIP3-induced cardiomyocyte death. Autophagy 2012, 8, 297–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, Q.; Li, R.; Hu, N.; Xin, T.; Zhu, P.; Hu, S.; Ma, S.; Zhu, H.; Ren, J.; Zhou, H. DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 2018, 14, 576–587. [Google Scholar] [CrossRef] [PubMed]
- Mao, S.; Tian, S.; Luo, X.; Zhou, M.; Cao, Z.; Li, J. Overexpression of PLK1 relieved the myocardial ischemia-reperfusion injury of rats through inducing the mitophagy and regulating the p-AMPK/FUNDC1 axis. Bioengineered 2021, 12, 2676–2687. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Yu, J. NR4A1 promotes cerebral ischemia reperfusion injury by repressing Mfn2-mediated mitophagy and inactivating the MAPK-ERK-CREB signaling pathway. Neurochem. Res. 2018, 43, 1963–1977. [Google Scholar] [CrossRef]
- Feng, Y.; Zhao, J.; Hou, H.; Zhang, H.; Jiao, Y.; Wang, J.; Wang, Y.; Sun, Y. WDR26 promotes mitophagy of cardiomyocytes induced by hypoxia through Parkin translocation. Acta Biochim. Biophys Sin. 2016, 48, 1075–1084. [Google Scholar] [CrossRef] [Green Version]
- Ji, W.; Wei, S.; Hao, P.; Xing, J.; Yuan, Q.; Wang, J.; Xu, F.; Chen, Y. Aldehyde dehydrogenase 2 has cardioprotective effects on myocardial ischaemia/reperfusion injury via suppressing mitophagy. Front. Pharmacol. 2016, 7, 101. [Google Scholar] [CrossRef] [Green Version]
- Yogalingam, G.; Hwang, S.; Ferreira, J.C.; Mochly-Rosen, D. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) phosphorylation by protein kinase Cdelta (PKCdelta) inhibits mitochondria elimination by lysosomal-like structures following ischemia and reoxygenation-induced injury. J. Biol. Chem. 2013, 288, 18947–18960. [Google Scholar] [CrossRef] [Green Version]
- Huang, C.; Andres, A.M.; Ratliff, E.P.; Hernandez, G.; Lee, P.; Gottlieb, R.A. Preconditioning involves selective mitophagy mediated by Parkin and p62/SQSTM1. PLoS ONE 2011, 6, e20975. [Google Scholar] [CrossRef]
- Wu, H.; Ye, M.; Liu, D.; Yang, J.; Ding, J.W.; Zhang, J.; Wang, X.A.; Dong, W.S.; Fan, Z.X.; Yang, J. UCP2 protect the heart from myocardial ischemia/reperfusion injury via induction of mitochondrial autophagy. J. Cell Biochem. 2019, 120, 15455–15466. [Google Scholar] [CrossRef]
- Xie, B.; Gao, X.; Huang, Y.; Zhang, Y.; Zhu, S. Remote Ischemic Postconditioning Inhibits Hippocampal Neuronal Apoptosis and Mitophagy After Cardiopulmonary Resuscitation in Rats. Shock 2021, 55, 74–82. [Google Scholar] [CrossRef]
- Huang, Y.; Gao, X.; Zhou, X.; Zhang, Y.; Tan, Z.; Zhu, S. Remote Ischemic Postconditioning Inhibited Mitophagy to Achieve Neuroprotective Effects in the Rat Model of Cardiac Arrest. Neurochem. Res. 2021, 46, 573–583. [Google Scholar] [CrossRef]
- Wen, H.; Li, L.; Zhan, L.; Zuo, Y.; Li, K.; Qiu, M.; Li, H.; Sun, W.; Xu, E. Hypoxic postconditioning promotes mitophagy against transient global cerebral ischemia via PINK1/Parkin-induced mitochondrial ubiquitination in adult rats. Cell Death Dis. 2021, 12, 630. [Google Scholar] [CrossRef] [PubMed]
- Bi, W.; Jia, J.; Pang, R.; Nie, C.; Han, J.; Ding, Z.; Liu, B.; Sheng, R.; Xu, J.; Zhang, J. Thyroid hormone postconditioning protects hearts from ischemia/reperfusion through reinforcing mitophagy. Biomed. Pharmacother. 2019, 118, 109220. [Google Scholar] [CrossRef]
- Wang, Y.; Lu, X.; Zhao, P.; Jiang, J.; Yao, L. Overexpression of PPENK reduces myocardial ischemia reperfusion injury by promoting mitophagy in rats. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2017, 33, 1335–1340. [Google Scholar] [PubMed]
- Yang, L.; Wu, J.; Xie, P.; Yu, J.; Li, X.; Wang, J.; Zheng, H. Sevoflurane postconditioning alleviates hypoxia-reoxygenation injury of cardiomyocytes by promoting mitochondrial autophagy through the HIF-1/BNIP3 signaling pathway. PeerJ 2019, 7, e7165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Correa, F.; Soto, V.; Zazueta, C. Mitochondrial permeability transition relevance for apoptotic triggering in the post-ischemic heart. Int. J. Biochem. Cell Biol. 2007, 39, 787–798. [Google Scholar] [CrossRef]
- Kroemer, G.; Galluzzi, L.; Vandenabeele, P.; Abrams, J.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V.; El-Deiry, W.S.; Golstein, P.; Green, D.R.; et al. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009, 16, 3–11. [Google Scholar] [CrossRef] [PubMed]
- Inserte, J.; Cardona, M.; Poncelas-Nozal, M.; Hernando, V.; Vilardosa, U.; Aluja, D.; Parra, V.M.; Sanchis, D.; Garcia-Dorado, D. Studies on the role of apoptosis after transient myocardial ischemia: Genetic deletion of the executioner caspases-3 and -7 does not limit infarct size and ventricular remodeling. Basic Res. Cardiol. 2016, 111, 18. [Google Scholar] [CrossRef]
- Chapman, J.G.; Magee, W.P.; Stukenbrok, H.A.; Beckius, G.E.; Milici, A.J.; Tracey, W.R. A novel nonpeptidic caspase-3/7 inhibitor, (S)-(+)-5-[1-(2-methoxymethylpyrrolidinyl)sulfonyl]isatin reduces myocardial ischemic injury. Eur.J. Pharmacol. 2002, 456, 59–68. [Google Scholar] [CrossRef]
- Kovacs, P.; Bak, I.; Szendrei, L.; Vecsernyes, M.; Varga, E.; Blasig, I.E.; Tosaki, A. Non-specific caspase inhibition reduces infarct size and improves post-ischaemic recovery in isolated ischaemic/reperfused rat hearts. Naunyn Schmiedebergs Arch. Pharmacol. 2001, 364, 501–507. [Google Scholar] [CrossRef]
- Ruetten, H.; Badorff, C.; Ihling, C.; Zeiher, A.M.; Dimmeler, S. Inhibition of caspase-3 improves contractile recovery of stunned myocardium, independent of apoptosis-inhibitory effects. J. Am. Coll. Cardiol. 2001, 38, 2063–2070. [Google Scholar] [CrossRef] [Green Version]
- Sun, T.; Ding, W.; Xu, T.; Ao, X.; Yu, T.; Li, M.; Liu, Y.; Zhang, X.; Hou, L.; Wang, J. Parkin regulates programmed necrosis and myocardial ischemia/reperfusion injury by targeting cyclophilin-D. Antioxid. Redox Signal. 2019, 31, 1177–1193. [Google Scholar] [CrossRef]
- Zhao, Z.Q.; Nakamura, M.; Wang, N.P.; Wilcox, J.N.; Shearer, S.; Ronson, R.S.; Guyton, R.A.; Vinten-Johansen, J. Reperfusion induces myocardial apoptotic cell death. Cardiovasc. Res. 2000, 45, 651–660. [Google Scholar] [CrossRef] [Green Version]
- Davidson, S.M.; Adameova, A.; Barile, L.; Cabrera-Fuentes, H.A.; Lazou, A.; Pagliaro, P.; Stenslokken, K.O.; Garcia-Dorado, D.; Action, E.-C.C. Mitochondrial and mitochondrial-independent pathways of myocardial cell death during ischaemia and reperfusion injury. J. Cell Mol. Med. 2020, 24, 3795–3806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parra-Flores, P.; Espitia-Corredor, J.; Espinoza-Perez, C.; Queirolo, C.; Ayala, P.; Bruggendieck, F.; Salas-Hernandez, A.; Pardo-Jimenez, V.; Diaz-Araya, G. Toll-like receptor 4 activation prevents rat cardiac fibroblast death induced by simulated ischemia/reperfusion. Front. Cardiovasc. Med. 2021, 8, 660197. [Google Scholar] [CrossRef]
- Roman-Anguiano, N.G.; Correa, F.; Cano-Martinez, A.; de la Pena-Diaz, A.; Zazueta, C. Cardioprotective effects of Prolame and SNAP are related with nitric oxide production and with diminution of caspases and calpain-1 activities in reperfused rat hearts. PeerJ 2019, 7, e7348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halestrap, A.P.; Davidson, A.M. Inhibition of Ca2+-induced large-amplitude swelling of liver and heart mitochondria by cyclosporin is probably caused by the inhibitor binding to mitochondrial-matrix peptidyl-prolyl cis-trans isomerase and preventing it interacting with the adenine nucleotide translocase. Biochem. J. 1990, 268, 153–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rück, A.; Dolder, M.; Wallimann, T.; Brdiczka, D. Reconstituted adenine nucleotide translocase forms a channel for small molecules comparable to the mitochondrial permeability transition pore. FEBS Lett. 1998, 426, 97–101. [Google Scholar] [CrossRef] [Green Version]
- Garcia, N.; Martinez-Abundis, E.; Pavon, N.; Chavez, E. Sodium inhibits permeability transition by decreasing potassium matrix content in rat kidney mitochondria. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2006, 144, 442–450. [Google Scholar] [CrossRef] [PubMed]
- Karch, J.; Bround, M.J.; Khalil, H.; Sargent, M.A.; Latchman, N.; Terada, N.; Peixoto, P.M.; Molkentin, J.D. Inhibition of mitochondrial permeability transition by deletion of the ANT family and CypD. Sci. Adv. 2019, 5, eaaw4597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonora, M.; Bononi, A.; De Marchi, E.; Giorgi, C.; Lebiedzinska, M.; Marchi, S.; Patergnani, S.; Rimessi, A.; Suski, J.M.; Wojtala, A.; et al. Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle 2013, 12, 674–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Westphal, D.; Kluck, R.M.; Dewson, G. Building blocks of the apoptotic pore: How Bax and Bak are activated and oligomerize during apoptosis. Cell Death Differ. 2014, 21, 196–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samali, A.; O’Mahoney, M.; Reeve, J.; Logue, S.; Szegezdi, E.; McMahon, J.; Fearnhead, H.O. Identification of an inhibitor of caspase activation from heart extracts; ATP blocks apoptosome formation. Apoptosis 2007, 12, 465–474. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Liang, P.; Jiang, B.; Tang, Y.; Lv, Q.; Hao, H.; Liu, Z.; Xiao, X. CARD9 inhibits mitochondria-dependent apoptosis of cardiomyocytes under oxidative stress via interacting with Apaf-1. Free Radic Biol. Med. 2019, 141, 172–181. [Google Scholar] [CrossRef] [PubMed]
- Degterev, A.; Huang, Z.; Boyce, M.; Li, Y.; Jagtap, P.; Mizushima, N.; Cuny, G.D.; Mitchison, T.J.; Moskowitz, M.A.; Yuan, J. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 2005, 1, 112–119. [Google Scholar] [CrossRef] [PubMed]
- Gurung, P.; Banskota, S.; Katila, N.; Gautam, J.; Kadayat, T.M.; Choi, D.Y.; Lee, E.S.; Jeong, T.C.; Kim, J.A. Ameliorating effect of TI-1-162, a hydroxyindenone derivative, against TNBS-induced rat colitis is mediated through suppression of RIP/ASK-1/MAPK signaling. Eur.J. Pharmacol. 2018, 827, 94–102. [Google Scholar] [CrossRef] [PubMed]
- He, M.X.; He, Y.W. A role for c-FLIP(L) in the regulation of apoptosis, autophagy, and necroptosis in T lymphocytes. Cell Death Differ. 2013, 20, 188–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pasparakis, M.; Vandenabeele, P. Necroptosis and its role in inflammation. Nature 2015, 517, 311–320. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Han, V.; Han, J. New components of the necroptotic pathway. Protein Cell 2012, 3, 811–817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tu, J.L.; Chen, W.P.; Cheng, Z.J.; Zhang, G.; Luo, Q.H.; Li, M.; Liu, X. EGb761 ameliorates cell necroptosis by attenuating RIP1-mediated mitochondrial dysfunction and ROS production in both in vivo and in vitro models of Alzheimer’s disease. Brain Res. 2020, 1736, 146730. [Google Scholar] [CrossRef] [PubMed]
- Bauer, T.M.; Murphy, E. Role of mitochondrial calcium and the permeability transition pore in regulating cell death. Circ. Res. 2020, 126, 280–293. [Google Scholar] [CrossRef]
- Gan, I.; Jiang, J.; Lian, D.; Huang, X.; Fuhrmann, B.; Liu, W.; Haig, A.; Jevnikar, A.M.; Zhang, Z.X. Mitochondrial permeability regulates cardiac endothelial cell necroptosis and cardiac allograft rejection. Am. J. Transplant. 2019, 19, 686–698. [Google Scholar] [CrossRef]
- Marshall, K.D.; Baines, C.P. Necroptosis: Is there a role for mitochondria? Front. Physiol. 2014, 5, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, P.; Hu, S.; Jin, Q.; Li, D.; Tian, F.; Toan, S.; Li, Y.; Zhou, H.; Chen, Y. Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol. 2018, 16, 157–168. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Ma, Q.; Toan, S.; Wang, J.; Zhou, H.; Liang, J. SERCA overexpression reduces reperfusion-mediated cardiac microvascular damage through inhibition of the calcium/MCU/mPTP/necroptosis signaling pathways. Redox Biol. 2020, 36, 101659. [Google Scholar] [CrossRef]
- Xiao, P.; Wang, C.; Li, J.; Su, H.; Yang, L.; Wu, P.; Lewno, M.T.; Liu, J.; Wang, X. COP9 signalosome suppresses RIPK1-RIPK3-mediated cardiomyocyte necroptosis in mice. Circ. Heart Fail. 2020, 13, e006996. [Google Scholar] [CrossRef] [PubMed]
- Minutoli, L.; Puzzolo, D.; Rinaldi, M.; Irrera, N.; Marini, H.; Arcoraci, V.; Bitto, A.; Crea, G.; Pisani, A.; Squadrito, F.; et al. ROS-mediated NLRP3 inflammasome activation in brain, heart, kidney, and testis ischemia/reperfusion injury. Oxid. Med. Cell.Longev. 2016, 2016, 2183026. [Google Scholar] [CrossRef]
- Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef]
- Ye, B.; Chen, X.; Dai, S.; Han, J.; Liang, X.; Lin, S.; Cai, X.; Huang, Z.; Huang, W. Emodin alleviates myocardial ischemia/reperfusion injury by inhibiting gasdermin D-mediated pyroptosis in cardiomyocytes. Drug Des. Devel. Ther. 2019, 13, 975–990. [Google Scholar] [CrossRef] [Green Version]
- Iyer, S.S.; He, Q.; Janczy, J.R.; Elliott, E.I.; Zhong, Z.; Olivier, A.K.; Sadler, J.J.; Knepper-Adrian, V.; Han, R.; Qiao, L.; et al. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity 2013, 39, 311–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nie, C.; Ding, X.; Rong, A.; Zheng, M.; Li, Z.; Pan, S.; Yang, W. Hydrogen gas inhalation alleviates myocardial ischemia-reperfusion injury by the inhibition of oxidative stress and NLRP3-mediated pyroptosis in rats. Life Sci. 2021, 272, 119248. [Google Scholar] [CrossRef] [PubMed]
- Griffiths, E.J.; Halestrap, A.P. Further evidence that cyclosporin A protects mitochondria from calcium overload by inhibiting a matrix peptidyl-prolyl cis-trans isomerase. Implications for the immunosuppressive and toxic effects of cyclosporin. Biochem. J. 1991, 274 Pt 2, 611–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cung, T.T.; Morel, O.; Cayla, G.; Rioufol, G.; Garcia-Dorado, D.; Angoulvant, D.; Bonnefoy-Cudraz, E.; Guerin, P.; Elbaz, M.; Delarche, N.; et al. Cyclosporine before PCI in patients with acute myocardial infarction. N. Engl. J. Med. 2015, 373, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
- Hurst, S.; Gonnot, F.; Dia, M.; Crola Da Silva, C.; Gomez, L.; Sheu, S.S. Phosphorylation of cyclophilin D at serine 191 regulates mitochondrial permeability transition pore opening and cell death after ischemia-reperfusion. Cell Death Dis. 2020, 11, 661. [Google Scholar] [CrossRef]
- Nguyen, T.T.; Stevens, M.V.; Kohr, M.; Steenbergen, C.; Sack, M.N.; Murphy, E. Cysteine 203 of cyclophilin D is critical for cyclophilin D activation of the mitochondrial permeability transition pore. J. Biol. Chem. 2011, 286, 40184–40192. [Google Scholar] [CrossRef] [Green Version]
- Amanakis, G.; Sun, J.; Fergusson, M.M.; McGinty, S.; Liu, C.; Molkentin, J.D.; Murphy, E. Cysteine 202 of cyclophilin D is a site of multiple post-translational modifications and plays a role in cardioprotection. Cardiovasc. Res. 2021, 117, 212–223. [Google Scholar] [CrossRef]
- Khaliulin, I.; Ascione, R.; Maslov, L.N.; Amal, H.; Suleiman, M.S. Preconditioning or Postconditioning with 8-Br-cAMP-AM Protects the Heart against Regional Ischemia and Reperfusion: A Role for Mitochondrial Permeability Transition. Cells 2021, 10, 1223. [Google Scholar] [CrossRef]
- Cai, X.; Yang, C.; Shao, L.; Zhu, H.; Wang, Y.; Huang, X.; Wang, S.; Hong, L. Targeting NOX 4 by petunidin improves anoxia/reoxygenation-induced myocardium injury. Eur.J. Pharmacol. 2020, 888, 173414. [Google Scholar] [CrossRef]
- Min, F.; Jia, X.J.; Gao, Q.; Niu, F.; Hu, Z.Y.; Han, Y.L.; Shi, H.J.; Yu, Y. Remote ischemic post-conditioning protects against myocardial ischemia/reperfusion injury by inhibiting the Rho-kinase signaling pathway. Exp. Ther. Med. 2020, 19, 99–106. [Google Scholar] [CrossRef] [Green Version]
- Tian, M.; Xie, Y.; Meng, Y.; Ma, W.; Tong, Z.; Yang, X.; Lai, S.; Zhou, Y.; He, M.; Liao, Z. Resveratrol protects cardiomyocytes against anoxia/reoxygenation via dephosphorylation of VDAC1 by Akt-GSK3 beta pathway. Eur. J. Pharmacol. 2019, 843, 80–87. [Google Scholar] [CrossRef]
- Cheng, X.; Hu, J.; Wang, Y.; Ye, H.; Li, X.; Gao, Q.; Li, Z. Effects of Dexmedetomidine Postconditioning on Myocardial Ischemia/Reperfusion Injury in Diabetic Rats: Role of the PI3K/Akt-Dependent Signaling Pathway. J. Diabetes Res. 2018, 2018, 3071959. [Google Scholar] [CrossRef]
- Yu, L.N.; Yu, J.; Zhang, F.J.; Yang, M.J.; Ding, T.T.; Wang, J.K.; He, W.; Fang, T.; Chen, G.; Yan, M. Sevoflurane postconditioning reduces myocardial reperfusion injury in rat isolated hearts via activation of PI3K/Akt signaling and modulation of Bcl-2 family proteins. J. Zhejiang Univ. Sci. B 2010, 11, 661–672. [Google Scholar] [CrossRef]
- Xi, J.; Wang, H.; Mueller, R.A.; Norfleet, E.A.; Xu, Z. Mechanism for resveratrol-induced cardioprotection against reperfusion injury involves glycogen synthase kinase 3beta and mitochondrial permeability transition pore. Eur. J. Pharmacol. 2009, 604, 111–116. [Google Scholar] [CrossRef] [Green Version]
- Liao, Z.; Liu, D.; Tang, L.; Yin, D.; Yin, S.; Lai, S.; Yao, J.; He, M. Long-term oral resveratrol intake provides nutritional preconditioning against myocardial ischemia/reperfusion injury: Involvement of VDAC1 downregulation. Mol. Nutr. Food Res. 2015, 59, 454–464. [Google Scholar] [CrossRef]
- Wang, J.; Zhou, H. Mitochondrial quality control mechanisms as molecular targets in cardiac ischemia-reperfusion injury. Acta Pharm. Sin. B 2020, 10, 1866–1879. [Google Scholar] [CrossRef]
- Tsai, C.F.; Su, H.H.; Chen, K.M.; Liao, J.M.; Yao, Y.T.; Chen, Y.H.; Wang, M.; Chu, Y.C.; Wang, Y.H.; Huang, S.S. Paeonol protects against myocardial ischemia/reperfusion-induced injury by mediating apoptosis and autophagy crosstalk. Front. Pharmacol. 2020, 11, 586498. [Google Scholar] [CrossRef]
- Smith, C.C.; Davidson, S.M.; Lim, S.Y.; Simpkin, J.C.; Hothersall, J.S.; Yellon, D.M. Necrostatin: A potentially novel cardioprotective agent? Cardiovasc. Drugs Ther. 2007, 21, 227–233. [Google Scholar] [CrossRef] [PubMed]
- She, L.; Tu, H.; Zhang, Y.Z.; Tang, L.J.; Li, N.S.; Ma, Q.L.; Liu, B.; Li, Q.; Luo, X.J.; Peng, J. Inhibition of phosphoglycerate mutase 5 reduces necroptosis in rat hearts following ischemia/reperfusion through suppression of dynamin-related protein 1. Cardiovasc. Drugs Ther. 2019, 33, 13–23. [Google Scholar] [CrossRef] [PubMed]
- Dmitriev, Y.V.; Minasian, S.M.; Demchenko, E.A.; Galagudza, M.M. Study of cardioprotective effects of necroptosis inhibitors on isolated rat heart subjected to global ischemia-reperfusion. Bull. Exp. Biol. Med. 2013, 155, 245–248. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, N.; Duprez, L.; Grootjans, S.; Cauwels, A.; Nerinckx, W.; DuHadaway, J.B.; Goossens, V.; Roelandt, R.; Van Hauwermeiren, F.; Libert, C.; et al. Necrostatin-1 analogues: Critical issues on the specificity, activity and in vivo use in experimental disease models. Cell Death Dis. 2012, 3, e437. [Google Scholar] [CrossRef] [Green Version]
- Horvath, C.; Young, M.; Jarabicova, I.; Kindernay, L.; Ferenczyova, K.; Ravingerova, T.; Lewis, M.; Suleiman, M.S.; Adameova, A. Inhibition of cardiac RIP3 mitigates early reperfusion injury and calcium-induced mitochondrial swelling without altering necroptotic signalling. Int. J. Mol. Sci. 2021, 22, 7983. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Yu, P.; Hua, F.; Hu, Y.; Xiao, F.; Liu, Q.; Huang, D.; Deng, F.; Wei, G.; Deng, W.; et al. Sevoflurane postconditioning reduces myocardial ischemia reperfusion injury-induced necroptosis by up-regulation of OGT-mediated O-GlcNAcylated RIPK3. Aging 2020, 12, 25452–25468. [Google Scholar] [CrossRef] [PubMed]
- Audrito, V.; Messana, V.G.; Deaglio, S. NAMPT and NAPRT: Two metabolic enzymes with key roles in inflammation. Front. Oncol. 2020, 10, 358. [Google Scholar] [CrossRef] [Green Version]
- Audia, J.P.; Yang, X.M.; Crockett, E.S.; Housley, N.; Haq, E.U.; O’Donnell, K.; Cohen, M.V.; Downey, J.M.; Alvarez, D.F. Caspase-1 inhibition by VX-765 administered at reperfusion in P2Y12 receptor antagonist-treated rats provides long-term reduction in myocardial infarct size and preservation of ventricular function. Basic Res. Cardiol. 2018, 113, 32. [Google Scholar] [CrossRef]
- Kawaguchi, M.; Takahashi, M.; Hata, T.; Kashima, Y.; Usui, F.; Morimoto, H.; Izawa, A.; Takahashi, Y.; Masumoto, J.; Koyama, J.; et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation 2011, 123, 594–604. [Google Scholar] [CrossRef] [Green Version]
- Armstrong, D.; Wildman, D.E. Extracellular vesicles and the promise of continuous liquid biopsies. J. Pathol. Transl. Med. 2018, 52, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Ratajczak, J.; Wysoczynski, M.; Hayek, F.; Janowska-Wieczorek, A.; Ratajczak, M.Z. Membrane-derived microvesicles: Important and underappreciated mediators of cell-to-cell communication. Leukemia 2006, 20, 1487–1495. [Google Scholar] [CrossRef]
- Fevrier, B.; Raposo, G. Exosomes: Endosomal-derived vesicles shipping extracellular messages. Curr. Opin. Cell Biol. 2004, 16, 415–421. [Google Scholar] [CrossRef] [PubMed]
- Kakarla, R.; Hur, J.; Kim, Y.J.; Kim, J.; Chwae, Y.J. Apoptotic cell-derived exosomes: Messages from dying cells. Exp. Mol. Med. 2020, 52, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kao, C.Y.; Papoutsakis, E.T. Extracellular vesicles: Exosomes, microparticles, their parts, and their targets to enable their biomanufacturing and clinical applications. Curr. Opin. Biotechnol. 2019, 60, 89–98. [Google Scholar] [CrossRef] [PubMed]
- Tetta, C.; Ghigo, E.; Silengo, L.; Deregibus, M.C.; Camussi, G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine 2013, 44, 11–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, D.S.; Kim, D.K.; Kim, Y.K.; Gho, Y.S. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics 2013, 13, 1554–1571. [Google Scholar] [CrossRef] [PubMed]
- Ikeda, G.; Santoso, M.R.; Tada, Y.; Li, A.M.; Vaskova, E.; Jung, J.H.; O’Brien, C.; Egan, E.; Ye, J.; Yang, P.C. Mitochondria-rich extracellular vesicles from autologous stem cell-derived cardiomyocytes restore energetics of ischemic myocardium. J. Am. Coll. Cardiol. 2021, 77, 1073–1088. [Google Scholar] [CrossRef] [PubMed]
- Haraszti, R.A.; Didiot, M.C.; Sapp, E.; Leszyk, J.; Shaffer, S.A.; Rockwell, H.E.; Gao, F.; Narain, N.R.; DiFiglia, M.; Kiebish, M.A.; et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J. Extracell. Vesicles 2016, 5, 32570. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.; Neupane, Y.R.; Lim, X.C.; Shekhani, R.; Czarny, B.; Wacker, M.G.; Pastorin, G.; Wang, J.W. Extracellular vesicles in cardiovascular disease. Adv. Clin. Chem. 2021, 103, 47–95. [Google Scholar] [CrossRef] [PubMed]
- Danielson, K.M.; Shah, R.; Yeri, A.; Liu, X.; Camacho Garcia, F.; Silverman, M.; Tanriverdi, K.; Das, A.; Xiao, C.; Jerosch-Herold, M.; et al. Plasma circulating extracellular RNAs in left ventricular remodeling post-myocardial infarction. EBioMedicine 2018, 32, 172–181. [Google Scholar] [CrossRef] [PubMed]
- Vicencio, J.M.; Yellon, D.M.; Sivaraman, V.; Das, D.; Boi-Doku, C.; Arjun, S.; Zheng, Y.; Riquelme, J.A.; Kearney, J.; Sharma, V.; et al. Plasma exosomes protect the myocardium from ischemia-reperfusion injury. J. Am. Coll. Cardiol. 2015, 65, 1525–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed]
- Yuan, Y.; Du, W.; Liu, J.; Ma, W.; Zhang, L.; Du, Z.; Cai, B. Stem cell-derived exosome in cardiovascular diseases: Macro roles of micro particles. Front. Pharmacol. 2018, 9, 547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, I.A.; Sanina, C.; Balkan, W.; Hare, J.M. Mesenchymal stem cells in cardiology. Methods Mol. Biol. 2016, 1416, 55–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, S.J.O.; Floriano, J.F.; Nicastro, L.; Emanueli, C.; Catapano, F. Novel applications of mesenchymal stem cell-derived exosomes for myocardial infarction therapeutics. Biomolecules 2020, 10, 707. [Google Scholar] [CrossRef]
- Colliva, A.; Braga, L.; Giacca, M.; Zacchigna, S. Endothelial cell-cardiomyocyte crosstalk in heart development and disease. J. Physiol. 2020, 598, 2923–2939. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Li, X.; Zhu, W.; Zhang, Y.; Hong, Y.; Liang, X.; Fan, B.; Zhao, H.; He, H.; Zhang, F. Exosomes from mesenchymal stem cells overexpressing MIF enhance myocardial repair. J. Cell Physiol. 2020, 235, 8010–8022. [Google Scholar] [CrossRef]
- Feng, Y.; Huang, W.; Wani, M.; Yu, X.; Ashraf, M. Ischemic preconditioning potentiates the protective effect of stem cells through secretion of exosomes by targeting Mecp2 via miR-22. PLoS ONE 2014, 9, e88685. [Google Scholar] [CrossRef]
- Gu, D.; Zou, X.; Ju, G.; Zhang, G.; Bao, E.; Zhu, Y. Mesenchymal stromal cells derived extracellular vesicles ameliorate acute renal ischemia reperfusion injury by inhibition of mitochondrial fission through miR-30. Stem Cells Int. 2016, 2016, 2093940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, X.; Wang, J.; Li, J.; Ma, C.; Chen, S.; Lei, W.; Yang, Y.; Liu, S.; Bihl, J.; Chen, C. Loading MiR-210 in endothelial progenitor cells derived exosomes boosts their beneficial effects on hypoxia/reoxygeneation-injured human endothelial cells via protecting mitochondrial function. Cell Physiol. Biochem. 2018, 46, 664–675. [Google Scholar] [CrossRef] [PubMed]
- Diao, H.; Liu, B.; Shi, Y.; Song, C.; Guo, Z.; Liu, N.; Song, X.; Lu, Y.; Lin, X.; Li, Z. MicroRNA-210 alleviates oxidative stress-associated cardiomyocyte apoptosis by regulating BNIP3. Biosci. Biotechnol. Biochem. 2017, 81, 1712–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; Jiang, M.; Deng, S.; Lu, J.; Huang, H.; Zhang, Y.; Gong, P.; Shen, X.; Ruan, H.; Jin, M.; et al. miR-93-5p-containing exosomes treatment attenuates acute myocardial infarction-induced myocardial damage. Mol. Ther. Nucleic Acids 2018, 11, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, Q.; Guo, D.; Liu, G.; Chen, G.; Hang, M.; Jin, M. Exosomes from MiR-126-overexpressing Adscs are therapeutic in relieving acute myocardial ischaemic injury. Cell Physiol. Biochem. 2017, 44, 2105–2116. [Google Scholar] [CrossRef]
- Pan, J.; Alimujiang, M.; Chen, Q.; Shi, H.; Luo, X. Exosomes derived from miR-146a-modified adipose-derived stem cells attenuate acute myocardial infarction-induced myocardial damage via downregulation of early growth response factor 1. J. Cell Biochem. 2019, 120, 4433–4443. [Google Scholar] [CrossRef]
- Crewe, C.; Funcke, J.B.; Li, S.; Joffin, N.; Gliniak, C.M.; Ghaben, A.L.; An, Y.A.; Sadek, H.A.; Gordillo, R.; Akgul, Y.; et al. Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. Cell Metab. 2021, 33, 1853–1868.e11. [Google Scholar] [CrossRef]
- Carr, C.A.; Stuckey, D.J.; Tan, J.J.; Tan, S.C.; Gomes, R.S.; Camelliti, P.; Messina, E.; Giacomello, A.; Ellison, G.M.; Clarke, K. Cardiosphere-derived cells improve function in the infarcted rat heart for at least 16 weeks-an MRI study. PLoS ONE 2011, 6, e25669. [Google Scholar] [CrossRef]
- Gallet, R.; Dawkins, J.; Valle, J.; Simsolo, E.; de Couto, G.; Middleton, R.; Tseliou, E.; Luthringer, D.; Kreke, M.; Smith, R.R.; et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur. Heart J. 2017, 38, 201–211. [Google Scholar] [CrossRef] [Green Version]
- Namazi, H.; Namazi, I.; Ghiasi, P.; Ansari, H.; Rajabi, S.; Hajizadeh-Saffar, E.; Aghdami, N.; Mohit, E. Exosomes secreted by normoxic and hypoxic cardiosphere-derived cells have anti-apoptotic effect. Iran. J. Pharm Res. 2018, 17, 377–385. [Google Scholar] [PubMed]
- Hergenreider, E.; Heydt, S.; Treguer, K.; Boettger, T.; Horrevoets, A.J.; Zeiher, A.M.; Scheffer, M.P.; Frangakis, A.S.; Yin, X.; Mayr, M.; et al. Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nat. Cell Biol. 2012, 14, 249–256. [Google Scholar] [CrossRef] [PubMed]
- Spannbauer, A.; Traxler, D.; Lukovic, D.; Zlabinger, K.; Winkler, J.; Gugerell, A.; Ferdinandy, P.; Hausenloy, D.J.; Pavo, N.; Emmert, M.Y.; et al. Effect of ischemic preconditioning and postconditioning on exosome-rich fraction microRNA levels, in relation with electrophysiological parameters and ventricular arrhythmia in experimental closed-chest reperfused myocardial infarction. Int. J. Mol. Sci. 2019, 20, 2140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, Y.; Vicencio, J.M.; Yellon, D.M.; Davidson, S.M. Exosomes released from endothelial cells are cardioprotective. Heart 2014, 100, 15885. [Google Scholar] [CrossRef]
- Hausenloy, D.J.; Tsang, A.; Yellon, D.M. The reperfusion injury salvage kinase pathway: A common target for both ischemic preconditioning and postconditioning. Trends Cardiovasc. Med. 2005, 15, 69–75. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Zhang, X. Ischaemic preconditioning-induced serum exosomes protect against myocardial ischaemia/reperfusion injury in rats by activating the PI3K/AKT signalling pathway. Cell Biochem. Funct. 2021, 39, 287–295. [Google Scholar] [CrossRef]
- Wang, Y.L.; Liu, M.; Shang, M.; Wang, Y.; Zhang, Q.; Wang, S.X.; Wei, S.; Zhang, K.W.; Liu, C.; Wu, Y.N.; et al. Effects of circulating microvesicles derived from myocardial ischemic preconditioning on myocardial ischemia/reperfusion injury in rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi 2016, 32, 97–101. [Google Scholar] [CrossRef]
- Liu, M.; Wang, Y.; Zhu, Q.; Zhao, J.; Wang, Y.; Shang, M.; Liu, M.; Wu, Y.; Song, J.; Liu, Y. Protective effects of circulating microvesicles derived from ischemic preconditioning on myocardial ischemia/reperfusion injury in rats by inhibiting endoplasmic reticulum stress. Apoptosis 2018, 23, 436–448. [Google Scholar] [CrossRef]
- Adams, J.M.; Cory, S. Bcl-2-regulated apoptosis: Mechanism and therapeutic potential. Curr. Opin. Immunol. 2007, 19, 488–496. [Google Scholar] [CrossRef] [Green Version]
- Sun, L.; Zhu, W.; Zhao, P.; Wang, Q.; Fan, B.; Zhu, Y.; Lu, Y.; Chen, Q.; Zhang, J.; Zhang, F. Long noncoding RNA UCA1 from hypoxia-conditioned hMSC-derived exosomes: A novel molecular target for cardioprotection through miR-873-5p/XIAP axis. Cell Death Dis. 2020, 11, 696. [Google Scholar] [CrossRef] [PubMed]
- Zhu, L.P.; Tian, T.; Wang, J.Y.; He, J.N.; Chen, T.; Pan, M.; Xu, L.; Zhang, H.X.; Qiu, X.T.; Li, C.C.; et al. Hypoxia-elicited mesenchymal stem cell-derived exosomes facilitates cardiac repair through miR-125b-mediated prevention of cell death in myocardial infarction. Theranostics 2018, 8, 6163–6177. [Google Scholar] [CrossRef] [PubMed]
- Luo, H.; Li, X.; Li, T.; Zhao, L.; He, J.; Zha, L.; Qi, Q.; Yu, Z. microRNA-423-3p exosomes derived from cardiac fibroblasts mediates the cardioprotective effects of ischaemic post-conditioning. Cardiovasc. Res. 2019, 115, 1189–1204. [Google Scholar] [CrossRef] [PubMed]
- Jeanneteau, J.; Hibert, P.; Martinez, M.C.; Tual-Chalot, S.; Tamareille, S.; Furber, A.; Andriantsitohaina, R.; Prunier, F. Microparticle release in remote ischemic conditioning mechanism. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H871–H877. [Google Scholar] [CrossRef] [Green Version]
- Giricz, Z.; Varga, Z.V.; Baranyai, T.; Sipos, P.; Paloczi, K.; Kittel, A.; Buzas, E.I.; Ferdinandy, P. Cardioprotection by remote ischemic preconditioning of the rat heart is mediated by extracellular vesicles. J. Mol. Cell. Cardiol. 2014, 68, 75–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Q.; Huang, M.; Wu, J.; Jiang, Q.; Zheng, X. Exosomes isolated from the plasma of remote ischemic conditioning rats improved cardiac function and angiogenesis after myocardial infarction through targeting Hsp70. Aging 2020, 12, 3682–3693. [Google Scholar] [CrossRef] [PubMed]
- Lassen, T.R.; Just, J.; Hjortbak, M.V.; Jespersen, N.R.; Stenz, K.T.; Gu, T.; Yan, Y.; Su, J.; Hansen, J.; Baek, R.; et al. Cardioprotection by remote ischemic conditioning is transferable by plasma and mediated by extracellular vesicles. Basic Res. Cardiol. 2021, 116, 16. [Google Scholar] [CrossRef]
- Abel, F.; Murke, F.; Gaida, M.; Garnier, N.; Ochsenfarth, C.; Theiss, C.; Thielmann, M.; Kleinbongard, P.; Giebel, B.; Peters, J.; et al. Extracellular vesicles isolated from patients undergoing remote ischemic preconditioning decrease hypoxia-evoked apoptosis of cardiomyoblasts after isoflurane but not propofol exposure. PLoS ONE 2020, 15, e0228948. [Google Scholar] [CrossRef]
- Yamaguchi, T.; Izumi, Y.; Nakamura, Y.; Yamazaki, T.; Shiota, M.; Sano, S.; Tanaka, M.; Osada-Oka, M.; Shimada, K.; Miura, K.; et al. Repeated remote ischemic conditioning attenuates left ventricular remodeling via exosome-mediated intercellular communication on chronic heart failure after myocardial infarction. Int. J. Cardiol. 2015, 178, 239–246. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Rohailla, S.; Gelber, N.; Rutka, J.; Sabah, N.; Gladstone, R.A.; Wei, C.; Hu, P.; Kharbanda, R.K.; Redington, A.N. MicroRNA-144 is a circulating effector of remote ischemic preconditioning. Basic Res. Cardiol. 2014, 109, 423. [Google Scholar] [CrossRef] [PubMed]
- Tao, L.; Huang, X.; Xu, M.; Yang, L.; Hua, F. MiR-144 protects the heart from hyperglycemia-induced injury by regulating mitochondrial biogenesis and cardiomyocyte apoptosis. FASEB J. 2020, 34, 2173–2197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Minghua, W.; Zhijian, G.; Chahua, H.; Qiang, L.; Minxuan, X.; Luqiao, W.; Weifang, Z.; Peng, L.; Biming, Z.; Lingling, Y.; et al. Plasma exosomes induced by remote ischaemic preconditioning attenuate myocardial ischaemia/reperfusion injury by transferring miR-24. Cell Death Dis. 2018, 9, 320. [Google Scholar] [CrossRef] [PubMed]
- Ma, F.; Liu, H.; Shen, Y.; Zhang, Y.; Pan, S. Platelet-derived microvesicles are involved in cardio-protective effects of remote preconditioning. Int. J. Clin. Exp. Pathol. 2015, 8, 10832–10839. [Google Scholar] [PubMed]
- Cadete, V.J.; Deschenes, S.; Cuillerier, A.; Brisebois, F.; Sugiura, A.; Vincent, A.; Turnbull, D.; Picard, M.; McBride, H.M.; Burelle, Y. Formation of mitochondrial-derived vesicles is an active and physiologically relevant mitochondrial quality control process in the cardiac system. J. Physiol. 2016, 594, 5343–5362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vincent, A.E.; Turnbull, D.M.; Eisner, V.; Hajnoczky, G.; Picard, M. Mitochondrial nanotunnels. Trends Cell Biol. 2017, 27, 787–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qin, Y.; Jiang, X.; Yang, Q.; Zhao, J.; Zhou, Q.; Zhou, Y. The functions, methods, and mobility of mitochondrial transfer between cells. Front. Oncol. 2021, 11, 672781. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
García-Niño, W.R.; Zazueta, C.; Buelna-Chontal, M.; Silva-Palacios, A. Mitochondrial Quality Control in Cardiac-Conditioning Strategies against Ischemia-Reperfusion Injury. Life 2021, 11, 1123. https://doi.org/10.3390/life11111123
García-Niño WR, Zazueta C, Buelna-Chontal M, Silva-Palacios A. Mitochondrial Quality Control in Cardiac-Conditioning Strategies against Ischemia-Reperfusion Injury. Life. 2021; 11(11):1123. https://doi.org/10.3390/life11111123
Chicago/Turabian StyleGarcía-Niño, Wylly Ramsés, Cecilia Zazueta, Mabel Buelna-Chontal, and Alejandro Silva-Palacios. 2021. "Mitochondrial Quality Control in Cardiac-Conditioning Strategies against Ischemia-Reperfusion Injury" Life 11, no. 11: 1123. https://doi.org/10.3390/life11111123
APA StyleGarcía-Niño, W. R., Zazueta, C., Buelna-Chontal, M., & Silva-Palacios, A. (2021). Mitochondrial Quality Control in Cardiac-Conditioning Strategies against Ischemia-Reperfusion Injury. Life, 11(11), 1123. https://doi.org/10.3390/life11111123