Targeting Autophagy for the Therapeutic Application of Histone Deacetylase Inhibitors in Ischemia/reperfusion Heart Injury.
Yingmei Zhang,Jun Ren
DOI: https://doi.org/10.1161/circulationaha.113.008115
IF: 37.8
2014-01-01
Circulation
Abstract:HomeCirculationVol. 129, No. 10Targeting Autophagy for the Therapeutic Application of Histone Deacetylase Inhibitors in Ischemia/Reperfusion Heart Injury Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBTargeting Autophagy for the Therapeutic Application of Histone Deacetylase Inhibitors in Ischemia/Reperfusion Heart Injury Yingmei Zhang, MD, PhD and Jun Ren, MD, PhD Yingmei ZhangYingmei Zhang From the Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie (Y.Z., J.R.); and Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China (Y.Z.). Search for more papers by this author and Jun RenJun Ren From the Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie (Y.Z., J.R.); and Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China (Y.Z.). Search for more papers by this author Originally published6 Jan 2014https://doi.org/10.1161/CIRCULATIONAHA.113.008115Circulation. 2014;129:1088–1091Other version(s) of this articleYou are viewing the most recent version of this article. Previous versions: March 11, 2014: Previous Version 1 Ischemic heart disease is a leading cause of morbidity and mortality in the United States and other parts of the world. Despite therapeutic breakthroughs over the past decades such as percutaneous coronary intervention, antiplatelet and antithrombotic therapies, and angioplasty, the prevalence of ischemic heart diseases remains extremely high and constitutes a devastating factor for heart failure.1,2 Among various therapeutic strategies for ischemic heart disease, enormous efforts have been made to limit ischemia/reperfusion (I/R) injury, which occurs when the ischemic myocardium is reperfused with oxygen and substrate-rich blood, which paradoxically worsens heart function.2 Ischemic myocardium, with nutrient and oxygen deprivation and buildup of reactive oxygen species (ROS), uses glycolysis as the primary source of metabolic energy. As a consequence, metabolic acidosis, hyperkalemia, and Ca2+ overload develop in cardiomyocytes after coronary artery occlusion, leading not only to cardiomyocyte apoptosis during the acute phase but also to delayed adverse myocardial remodeling, which further compromises cardiac function.2 Therefore, limiting I/R-induced myocardial ROS accumulation and apoptosis benefits both short- and long-term survival and quality of life. Although the mechanism responsible for I/R-induced cardiac abnormalities has been focused largely on necrosis and type I (apoptotic) programmed cell death,2 an intriguing and provocative paradigm has emerged recently that highlights a unique role for dysregulated macroautophagy (hereafter referred to as autophagy) in the heart that may render cardiomyocytes more prone to I/R injury and long-term postinfarction cardiac remodeling.1,3 It has been perceived that autophagy induced by ischemic preconditioning is essential for cardioprotection. To this end, new and innovative strategies to maintain or restore myocardial autophagy homeostasis and its attendant, cardiomyocyte survival, have been the subject of intensive investigation.Article see p 1139The Janus-Faced Role of Autophagy Induction in I/R InjuryAutophagy is a tightly regulated, lysosome-dependent catabolic process responsible for turnover of long-lived proteins and intracellular structures that are damaged or malfunctioning.4,5 The evolutionally conserved bulk degradation process is turned on when cells experience stress, including nutrient and energy deprivation. The autophagic pathway consists of 4 distinct but consecutive steps: initiation, formation of autophagosomes (ie, the double-membrane structures that encircle cargo of damaged cytosolic constituents), generation of autophagolysosomes via docking and fusion with lysosomes, and final degradation of sequestered cargo. Sequestration of cytoplasmic cargo such as long-lived proteins, damaged organelles, and protein aggregates into the double-membrane vesicle autophagosomes occurs before fusion with lysosomes for degradation of its contents by acidic hydrolases. Although physiological levels of autophagy are essential for mitochondrial function, cell survival, and cell function, excessive activation of autophagy can be detrimental, leading ultimately to autophagic cell death. Recent findings identified an important role for autophagy in the pathogenesis of human diseases, in particular heart diseases, implicating the therapeutic potential of autophagy regulation against heart anomalies.4,5 In light of the indispensable role of autophagy for cardiac homeostasis, recent attention has focused on understanding the role of autophagy regulation in I/R injury. It has been demonstrated that autophagy seems to play a paradoxical role in I/R injury. In ischemia, induction of autophagy via AMP kinase is protective, whereas reperfusion stimulates autophagy with Beclin-1 upregulation to compromise cardiac cell survival and function.3,6 This is in line with the observation that preischemic autophagy induction (eg, by chloramphenicol succinate) limits myocardial infarction in swine hearts. In addition, cardioprotection of delayed preconditioning by sevoflurane, a general anesthetic, is mediated by upregulation of autophagy. On the other hand, autophagy inhibition has been demonstrated to be responsible for the protective properties of mitochondrial aldehyde dehydrogenase 2 and chemokine CXCL16 against reperfusion injury.3 This dual regulatory paradigm of autophagy in the ischemia and reperfusion phases may underscore the homeostatic and drug intervention machinery for I/R heart injury. Further evidence indicates that I/R injury impairs autophagosome clearance (autophagy flux) mediated in part through an ROS-induced decline in lysosome-associated membrane protein-2 and upregulation of the autophagy initiation protein Beclin-1, leading to the ultimate cardiomyocyte death.7 Recently, a number of pharmaceutical therapies targeting I/R injury have been designed to orchestrate multiple protein complexes and signaling pathways in autophagy. For instance, sevoflurane has been shown to offer cardioprotection through ROS-mediated upregulation of autophagy in I/R.8 On the contrary, in vitro evidence suggested that α-lipoic acid protects H9C2 myoblasts against hypoxia/reoxygenation injury through suppression of autophagy.9 The precise role of autophagy regulation contributing to cell survival and death in ischemic hearts remains controversial. In this issue of Circulation, Xie and colleagues10 report that suberoylanilide hydroxamic acid (SAHA; vorinostat), a histone deacetylase (HDAC) inhibitor approved by the Food and Drug Administration for cancer treatment, attenuated myocardial reperfusion injury in rabbits. These results revealed that SAHA reduced infarct size and partially rescued systolic function when administered either before surgery or at the time of reperfusion. SAHA was found to facilitate autophagic flux in the infarct border zone in rabbit myocardium and in mice harboring an red fluorescent protein-green fluorescent protein-LC3 transgene. In cultured myocytes subjected to I/R, SAHA overtly alleviated cell death, the effect of which was correlated with increased autophagy. The permissive role of autophagy in SAHA-related beneficial effects was consolidated by the mitigation of SAHA efficacy through RNAi knockdown of autophagy genes Atg7 and Atg5. These findings have great clinical relevance because the plasma SAHA levels were similar to those achieved in cancer patients.10 This work has unveiled a new paradigm for the clinical utility of HDAC inhibitors and autophagy regulators in ischemic heart diseases. A plethora of studies have demonstrated proven cardioprotective benefits of HDAC inhibitors in models of myocardial stress, including cardiac hypertrophy, I/R, and heart failure.11–13 In particular, trichostatin A, a class I and II HDAC inhibitor structurally homologous to SAHA, reduces myocardial infarct size up to 50%.14 HDAC inhibition caused a dramatic increase in phosphorylation of p38 and p38 activity in the heart.14 Of note, HDAC inhibitors can be delivered as late as 1 hour after an ischemic insult and can achieve a similar degree of infarct size reduction using pretreatment, indicating the suitability of HDAC inhibitors to treat myocardial infarction at the time of percutaneous coronary intervention. Although discrepancies exist in disease mechanisms in animal models relative to the human case, these data clearly show that facilitated autophagy is required for HDAC inhibition–induced protection against I/R injury.10 Given the recent therapeutic promises using HDAC inhibitors in ischemic and hypertrophic heart diseases,11,12,15,16 the finding that SAHA rescues I/R heart injury through modulating autophagy flux is of great clinical importance. Interestingly, a number of cardioprotective agents such as the angiotensin II receptor blocker valsartan may also elicit protection against I/R injury through autophagy induction. Valsartan preconditioning is believed to facilitate autophagy induction via an Akt/mammalian target of rapamycin/S6K–mediated mechanism, although the underlying molecular mechanism behind SAHA-induced autophagy flux remains unclear at this time.The New Horizon of HDAC Inhibitors in Heart DiseasesHistone acetylation participates in the regulation of transcription by promoting a more relaxed chromatin structure necessary for transcriptional activation. Many proteins are regulated by reversible acetylation of ε-amino groups on lysine residues. Reversible protein acetylation is controlled by enzymes that either attach (histone acetyltransferases) or remove (HDACs) acetyl groups.13 With the removal of acetyl groups from an ε-N-acetyl lysine amino acid on a histone to restore the positive charge to lysine residues, HDAC proteins may also be referred to as lysine deacetylases to more precisely recapitulate their function rather than their targets.16–18 Small-molecule HDAC inhibitors, acting specifically or broadly on 1 or more of the 4 HDACs and on nonhistone targets, are currently being tested for oncological indications.16,18Gene deletion and overexpression studies have unveiled important functions of HDACs in a number of nononcological settings such as respiratory stress, inflammation, and cardiac remodeling, apoptosis, necrosis, metabolism, contractility, and fibrosis.11,15–18 HDACs have received attention as a potential new target for the treatment of heart diseases. Small-molecule HDAC inhibitors have demonstrated efficacy in animal models of heart failure.13 For instance, MPT0E014, a novel HDAC inhibitor, displayed remarkable HDAC 1, 2, and 6 isoenzyme suppressive properties, improved cardiac contractility, and retarded cardiac remodeling in isoproterenol-induced dilated cardiomyopathy.19 Several explanations for MPT0E014-induced beneficial effects have been suggested, including inhibition of migration and proliferation of cardiac fibroblasts, as well as cardiac fibrosis, decreased levels of atrial natriuretic peptide, angiotensin II type I receptor, transforming growth factor-β, and Ca2+/calmodulin-dependent protein kinase IIδ.19 Along the same lines, HDAC inhibition may retard cardiac remodeling and improve ventricular dysfunction caused by pressure overload.13,20 Cao and colleagues15 recently reported that the prototypical HDAC inhibitor trichostatin A attenuated both load- and agonist-induced hypertrophic myocardial growth through the inhibition of autophagy rather than facilitating autophagy as reported for SAHA.10 Although these findings appear to be contradictory, it is imperative that the HDAC inhibitors strive to restore autophagy homeostasis through inhibition of excessive autophagy flux under pressure overload15 or reactivation of autophagy flux beneficial to infarct boarder zone10 (Figure). Although Xie and colleagues did not examine ROS production in ischemic hearts treated with or without SAHA, it is possible that oxidative stress may mediate SAHA-related beneficial stimulation of autophagy flux. NADPH oxidase–mediated oxidative stress is well known to stimulate autophagic flux during myocardial I/R. HDAC inhibition has been shown to promote the accumulation of ROS. However, how ROS regulate autophagy is not fully understood, although it is possible that oxidative modification of transcription factors affects the expression of autophagy proteins. In addition, ROS may directly regulate the formation of autophagosomes. The autophagy gene Atg4 enables the conversion of LC3-I to lipidated LC3-II, its insertion into autophagosomes, and the recycling of LC3-II after autophagosome-lysosome fusion. It has been demonstrated that Atg4 is subject to oxidation, thus resulting in LC3-II accumulation and autophagosome formation. I/R injury–induced ROS production has been demonstrated to promote nuclear factor-κB activity and to accentuate myocardial injury via the activation of Beclin-1–mediated autophagy. On the other side of the coin, a number of autophagy regulatory proteins such as the mitochondrial autophagy protein Parkin possess multiple conserved cysteine residues that are susceptible to modification by oxidative and nitrosative stress, leading to aberrant modification and subsequently inhibition of autophagy. Considering the current controversy on the role of adaptive or maladaptive autophagy in cardiac pathology,5 it is pertinent to better understand conditions in which autophagy should be inhibited or activated not only to best preserve cardiac homeostasis but also to optimize drug therapy (such as the drug efficacy and resistance for HDAC inhibitors). Furthermore, it is noteworthy that the overall efficacy of HDAC inhibition is determined by the pleiotropic salutary actions in various cell types and pathophysiological processes, many of which (eg, anti-inflammation and NADPH oxidase) may be autophagy independent.HDACs are pivotal epigenetic regulatory enzymes with the ability to deacetylate nucleosome histones and nonhistone proteins and to elicit significant pathological effects in tumor growth and cardiovascular diseases. Two HDAC inhibitors, vorinostat and romidespin, have been approved by the Food and Drug Administration for cancer therapy. Although small-molecule HDAC inhibitors have displayed an unforeseen potential in the management of heart diseases, the broad-spectrum, “pan” HDAC inhibition is associated with toxicities such as thrombocytopenia, neutropenia, diarrhea, nausea, vomiting, and fatigue. Experts in the field remain skeptical of the prospects of translating these preclinical findings to the clinical setting. With the improved safety profiles of newly identified small-molecule HDAC inhibitors selectively targeting 1 or a small subset of the 18 human HDACs, identifying the HDAC isoform(s) that govern pathological cardiac remodeling and understanding the roles of acetylation/deacetylation in the regulation of autophagy and autophagy-independent cellular processes and the underlying molecular mechanisms should be the logical next steps for the advancement of safer HDAC inhibitors into clinical trials for heart diseases, particularly ischemic heart disease.18Download figureDownload PowerPointFigure. Role of autophagy in histone deacetylase (HDAC) inhibition–elicited protection against cardiac pathologies. The possible role of autophagy regulation in the cardioprotective effect of HDAC inhibitors in ischemia/reperfusion (I/R) heart injury is depicted. Stimulus (load and agonist)-induced cardiac hypertrophy is also displayed for comparison. HDAC inhibitors (eg, suberoylanilide hydroxamic acid) restore autophagy homeostasis in ischemic hearts possibly through reactivation of autophagy flux deemed beneficial to infarct boarder zone. In cardiac hypertrophy, HDAC inhibitors may suppress excessive autophagy flux to maintain cardiac autophagy homeostasis. Dashed lines represent likely, although unproven, cell signaling mechanisms. AC indicates acetylation; and ROS, reactive oxygen species.Sources of FundingWorks in the authors’ laboratories is supported by NIGMS8P20 GM103432 and National Natural Science Foundation of China81370195.DisclosuresNone.FootnotesThe opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.Correspondence to Jun Ren, MD, PhD, FAHA, Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071. E-mail [email protected]References1. Przyklenk K, Dong Y, Undyala VV, Whittaker P. Autophagy as a therapeutic target for ischaemia/reperfusion injury? Concepts, controversies, and challenges.Cardiovasc Res. 2012; 94:197–205.CrossrefMedlineGoogle Scholar2. Jennings RB. Historical perspective on the pathology of myocardial ischemia/reperfusion injury.Circ Res. 2013; 113:428–438.LinkGoogle Scholar3. Ma H, Guo R, Yu L, Zhang Y, Ren J. Aldehyde dehydrogenase 2 (ALDH2) rescues myocardial ischaemia/reperfusion injury: role of autophagy paradox and toxic aldehyde.Eur Heart J. 2011; 32:1025–1038.CrossrefMedlineGoogle Scholar4. Choi AM, Ryter SW, Levine B. Autophagy in human health and disease.N Engl J Med. 2013; 368:651–662.CrossrefMedlineGoogle Scholar5. Lavandero S, Troncoso R, Rothermel BA, Martinet W, Sadoshima J, Hill JA. Cardiovascular autophagy: concepts, controversies and perspectives.Autophagy. 2013; 9:1455–1466.CrossrefMedlineGoogle Scholar6. Hariharan N, Zhai P, Sadoshima J. Oxidative stress stimulates autophagic flux during ischemia/reperfusion.Antioxid Redox Signal. 2011; 14:2179–2190.CrossrefMedlineGoogle Scholar7. Ma X, Liu H, Foyil SR, Godar RJ, Weinheimer CJ, Hill JA, Diwan A. Impaired autophagosome clearance contributes to cardiomyocyte death in ischemia/reperfusion injury.Circulation. 2012; 125:3170–3181.LinkGoogle Scholar8. Shiomi M, Miyamae M, Takemura G, Kaneda K, Inamura Y, Onishi A, Koshinuma S, Momota Y, Minami T, Figueredo VM. Sevoflurane induces cardioprotection through reactive oxygen species-mediated upregulation of autophagy in isolated guinea pig hearts.J Anesth. December 12, 2013. doi:10.1007/ss00540-013-1755-9. http://link.springer.com/article/10.1007%2Fs00540-013-1755-9. Accessed December 18, 2013.MedlineGoogle Scholar9. Cao X, Chen A, Yang P, Song X, Liu Y, Li Z, Wang X, Wang L, Li Y. Alpha-lipoic acid protects cardiomyocytes against hypoxia/reoxygenation injury by inhibiting autophagy.Biochem Biophys Res Commun. 2013; 441:935–940.CrossrefMedlineGoogle Scholar10. Xie M, Kong Y, Tan W, May H, Battiprolu PK, Pedrozo Z, Wang ZV, Morales C, Luo X, Cho G, Jiang N, Jessen ME, Warner JJ, Lavandero S, Gillette TG, Turer AT, Hill JA. Histone deacetylase inhibition blunts ischemia/reperfusion injury by inducing cardiomyocyte autophagy.Circulation. 2014; 129:1139–1151.LinkGoogle Scholar11. Granger A, Abdullah I, Huebner F, Stout A, Wang T, Huebner T, Epstein JA, Gruber PJ. Histone deacetylase inhibition reduces myocardial ischemia-reperfusion injury in mice.FASEB J. 2008; 22:3549–3560.CrossrefMedlineGoogle Scholar12. Kong Y, Tannous P, Lu G, Berenji K, Rothermel BA, Olson EN, Hill JA. Suppression of class I and II histone deacetylases blunts pressure-overload cardiac hypertrophy.Circulation. 2006; 113:2579–2588.LinkGoogle Scholar13. Xie M, Hill JA. HDAC-dependent ventricular remodeling.Trends Cardiovasc Med. 2013; 23:229–235.CrossrefMedlineGoogle Scholar14. Zhao TC, Cheng G, Zhang LX, Tseng YT, Padbury JF. Inhibition of histone deacetylases triggers pharmacologic preconditioning effects against myocardial ischemic injury.Cardiovasc Res. 2007; 76:473–481.CrossrefMedlineGoogle Scholar15. Cao DJ, Wang ZV, Battiprolu PK, Jiang N, Morales CR, Kong Y, Rothermel BA, Gillette TG, Hill JA. Histone deacetylase (HDAC) inhibitors attenuate cardiac hypertrophy by suppressing autophagy.Proc Natl Acad Sci U S A. 2011; 108:4123–4128.CrossrefMedlineGoogle Scholar16. McKinsey TA. Therapeutic potential for HDAC inhibitors in the heart.Annu Rev Pharmacol Toxicol. 2012; 52:303–319.CrossrefMedlineGoogle Scholar17. Bush EW, McKinsey TA. Protein acetylation in the cardiorenal axis: the promise of histone deacetylase inhibitors.Circ Res. 2010; 106:272–284.LinkGoogle Scholar18. McKinsey TA. Isoform-selective HDAC inhibitors: closing in on translational medicine for the heart.J Mol Cell Cardiol. 2011; 51:491–496.CrossrefMedlineGoogle Scholar19. Kao YH, Liou JP, Chung CC, Lien GS, Kuo CC, Chen SA, Chen YJ. Histone deacetylase inhibition improved cardiac functions with direct antifibrotic activity in heart failure.Int J Cardiol. 2013; 168:4178–4183.CrossrefMedlineGoogle Scholar20. Tao H, Shi KH, Yang JJ, Huang C, Zhan HY, Li J. Histone deacetylases in cardiac fibrosis: current perspectives for therapy.Cell Signal. 2013; 26:521–527.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Xu C, Liu Y, Yang J, Zhai M, Fan Z, Qiao R, Jin P and Yang L (2022) Effects of berbamine against myocardial ischemia/reperfusion injury: Activation of the 5' adenosine monophosphate‐activated protein kinase/nuclear factor erythroid 2‐related factor pathway and changes in the mitochondrial state , BioFactors, 10.1002/biof.1820, 48:3, (651-664), Online publication date: 1-May-2022. Guo C, Zhang L, Gao Y, Sun J, Fan L, Bai Y, Zhang J, Naren G, Yang J, Li L and Pichini S (2021) Cox-2 Antagonizes the Protective Effect of Sevoflurane on Hypoxia/Reoxygenation-Induced Cardiomyocyte Apoptosis through Inhibiting the Akt Pathway, Disease Markers, 10.1155/2021/4114593, 2021, (1-6), Online publication date: 7-Dec-2021. Han R, Huang H, Han H, Chen H, Zeng F, Xie X, Liu D, Cai Y, Zhang L, Liu X, Xia Z and Tang J (2021) Propofol postconditioning ameliorates hypoxia/reoxygenation induced H9c2 cell apoptosis and autophagy via upregulating forkhead transcription factors under hyperglycemia, Military Medical Research, 10.1186/s40779-021-00353-0, 8:1, Online publication date: 1-Dec-2021. Chen X, Xie Q, Zhu Y, Xu J, Lin G, Liu S, Su Z, Lai X, Li Q, Xie J and Yang X (2021) Cardio-protective effect of tetrahydrocurcumin, the primary hydrogenated metabolite of curcumin in vivo and in vitro: Induction of apoptosis and autophagy via PI3K/AKT/mTOR pathways, European Journal of Pharmacology, 10.1016/j.ejphar.2021.174495, 911, (174495), Online publication date: 1-Nov-2021. Ren J (2021) Cardiovascular Medicine in the Era of COVID-19 Pandemics, Cardiology Plus, 10.4103/2470-7511.334404, 6:4, (199-201), Online publication date: 1-Oct-2021. Cho K and Kim G (2021) Selective striatal cell loss is ameliorated by regulated autophagy of the cortex, Life Sciences, 10.1016/j.lfs.2021.119822, 282, (119822), Online publication date: 1-Oct-2021. Ren J, Wang X and Zhang Y (2021) Editorial: New Drug Targets for Proteotoxicity in Cardiometabolic Diseases, Frontiers in Physiology, 10.3389/fphys.2021.745296, 12 Naseroleslami M, Rakhshan K, Aboutaleb N and Souri F (2021) Lavender Oil Attenuates Myocardial Ischemia/Reperfusion Injury Through Inhibition of Autophagy and Stimulation of Angiogenesis, Iranian Journal of Science and Technology, Transactions A: Science, 10.1007/s40995-021-01123-2, 45:4, (1201-1209), Online publication date: 1-Aug-2021. Xie M, Cho G, Kong Y, Li D, Altamirano F, Luo X, Morales C, Jiang N, Schiattarella G, May H, Medina J, Shelton J, Ferdous A, Gillette T and Hill J (2021) Activation of Autophagic Flux Blunts Cardiac Ischemia/Reperfusion Injury, Circulation Research, 129:3, (435-450), Online publication date: 23-Jul-2021. Xu C, Wang J, Fan Z, Zhang S, Qiao R, Liu Y, Yang J, Yang L and Wang H (2021) Cardioprotective effects of melatonin against myocardial ischaemia/reperfusion injury: Activation of AMPK/Nrf2 pathway, Journal of Cellular and Molecular Medicine, 10.1111/jcmm.16691, 25:13, (6455-6459), Online publication date: 1-Jul-2021. Zhang J and Zhang X (2020) Ischaemic preconditioning‐induced serum exosomes protect against myocardial ischaemia/reperfusion injury in rats by activating the PI3K / AKT signalling pathway , Cell Biochemistry and Function, 10.1002/cbf.3578, 39:2, (287-295), Online publication date: 1-Mar-2021. Yang M, Zhang Y and Ren J (2020) Acetylation in cardiovascular diseases: Molecular mechanisms and clinical implications, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 10.1016/j.bbadis.2020.165836, 1866:10, (165836), Online publication date: 1-Oct-2020. Tian H, Liu S, Ren J, Lee J, Wang R and Chen P (2020) Role of Histone Deacetylases in Skeletal Muscle Physiology and Systemic Energy Homeostasis: Implications for Metabolic Diseases and Therapy, Frontiers in Physiology, 10.3389/fphys.2020.00949, 11 Naseroleslami M, Sharifi M, Rakhshan K, Mokhtari B and Aboutaleb N (2020) Nesfatin-1 attenuates injury in a rat model of myocardial infarction by targeting autophagy, inflammation, and apoptosis, Archives of Physiology and Biochemistry, 10.1080/13813455.2020.1802486, (1-9) Wu N, Tian H, Chen P, Wang D, Ren J and Zhang Y (2019) Physical Exercise and Selective Autophagy: Benefit and Risk on Cardiovascular Health, Cells, 10.3390/cells8111436, 8:11, (1436) Duan J, Chen Z, Wu Y, Zhu B, Yang L and Yang C (2019) Metabolic remodeling induced by mitokines in heart failure, Aging, 10.18632/aging.102247, 11:17, (7307-7327), Online publication date: 9-Sep-2019. Chen X, Wang L, Deng Y, Li X, Li G, Zhou J, Cheng D, Yang Y, Yang Q, Chen G and Wang G (2019) Inhibition of Autophagy Prolongs Recipient Survival Through Promoting CD8+ T Cell Apoptosis in a Rat Liver Transplantation Model, Frontiers in Immunology, 10.3389/fimmu.2019.01356, 10 Chen P, Shang A, Yang J and Wang W (2018) microRNA‐874 inhibition targeting STAT3 protects the heart from ischemia–reperfusion injury by attenuating cardiomyocyte apoptosis in a mouse model, Journal of Cellular Physiology, 10.1002/jcp.27398, 234:5, (6182-6193), Online publication date: 1-May-2019. Li Y, Zhao K, Zong P, Fu H, Zheng Y, Bao D, Yin Y, Chen Q, Lu L, Dai Y, Hou D and Kong X (2019) CD47 deficiency protects cardiomyocytes against hypoxia/reoxygenation injury by rescuing autophagic clearance, Molecular Medicine Reports, 10.3892/mmr.2019.10199 Wang R, Lin J and Bagchi R (2019) Novel molecular therapeutic targets in cardiac fibrosis: a brief overview, Canadian Journal of Physiology and Pharmacology, 10.1139/cjpp-2018-0430, 97:4, (246-256), Online publication date: 1-Apr-2019. Su J, Xu T, Jiang G, Hou M, Liang M, Cheng H and Li Q (2019) Gambogenic acid triggers apoptosis in human nasopharyngeal carcinoma CNE-2Z cells by activating volume-sensitive outwardly rectifying chloride channel, Fitoterapia, 10.1016/j.fitote.2019.01.002, 133, (150-158), Online publication date: 1-Mar-2019. Shao J, Miao C, Geng Z, Gu M, Wu Y and Li Q (2019) Effect of eNOS on Ischemic Postconditioning-Induced Autophagy against Ischemia/Reperfusion Injury in Mice, BioMed Research International, 10.1155/2019/5201014, 2019, (1-11), Online publication date: 10-Feb-2019. Ren Z, Xiao W, Zeng Y, Liu M, Li G, Tang Z, Qu S, Hao Y, Yuan H and Jiang Z (2019) Fibroblast growth factor-21 alleviates hypoxia/reoxygenation injury in H9c2 cardiomyocytes by promoting autophagic flux, International Journal of Molecular Medicine, 10.3892/ijmm.2019.4071 Cheng Z, Zhang M, Hu J, Lin J, Feng X, Wang S, Wang T, Gao E, Wang H and Sun D (2018) Mst1 knockout enhances cardiomyocyte autophagic flux to alleviate angiotensin II-induced cardiac injury independent of angiotensin II receptors, Journal of Molecular and Cellular Cardiology, 10.1016/j.yjmcc.2018.08.028, 125, (117-128), Online publication date: 1-Dec-2018. Chang C, Li S, Chen Y, Huang S, Chen S and Chen Y (2018) Histone deacetylase inhibition attenuates atrial arrhythmogenesis in sterile pericarditis, Translational Research, 10.1016/j.trsl.2018.06.002, 200, (54-64), Online publication date: 1-Oct-2018. Gyongyosi A, Terraneo L, Bianciardi P, Tosaki A, Lekli I and Samaja M (2018) The Impact of Moderate Chronic Hypoxia and Hyperoxia on the Level of Apoptotic and Autophagic Proteins in Myocardial Tissue, Oxidative Medicine and Cellular Longevity, 10.1155/2018/5786742, 2018, (1-12), Online publication date: 16-Aug-2018. Liu Y, Sun C, Xue F, Yang G, Li H, Liu Q and Liao X (2018) Effect of Autophagy Inhibition on the Protection of Ischemia Preconditioning against Myocardial Ischemia/Reperfusion Injury in Diabetic Rats, Chinese Medical Journal, 10.4103/0366-6999.235867, 131:14, (1702-1709), Online publication date: 20-Jul-2018. Shao H, Yang L, Wang L, Tang B, Wang J and Li Q (2018) MicroRNA-34a protects myocardial cells against ischemia–reperfusion injury through inhibiting autophagy via regulating TNFα expression, Biochemistry and Cell Biology, 10.1139/bcb-2016-0158, 96:3, (349-354), Online publication date: 1-Jun-2018. Guo W, Liu X, Li J, Shen Y, Zhou Z, Wang M, Xie Y, Feng X, Wang L and Wu X (2018) Prdx1 alleviates cardiomyocyte apoptosis through ROS-activated MAPK pathway during myocardial ischemia/reperfusion injury, International Journal of Biological Macromolecules, 10.1016/j.ijbiomac.2018.02.009, 112, (608-615), Online publication date: 1-Jun-2018. Wright L, Herr D, Brown S, Kasiganesan H and Menick D (2018)(2018)(2018)(2018) Angiokine Wisp-1 is increased in myocardial infarction and regulates cardiac endothelial signaling, JCI Insight, 10.1172/jci.insight.95824, 3:4, Online publication date: 22-Feb-2018., Online publication date: 22-Feb-2018., Online publication date: 22-Feb-2018., Online publication date: 22-Feb-2018. Jiang X, Liu G, Lei H, Li Z, Feng Q and Huang W (2017) Activation of transient receptor potential vanilloid 1 protects the heart against apoptosis in ischemia/reperfusion injury through upregulating the PI3K/Akt signaling pathway, International Journal of Molecular Medicine, 10.3892/ijmm.2017.3338 Rasekhi M, Soleimani M, Bakhshandeh B and Sadeghizadeh M (2017) A novel protocol to provide a suitable cardiac model from induced pluripotent stem cells, Biologicals, 10.1016/j.biologicals.2017.09.003, 50, (42-48), Online publication date: 1-Nov-2017. Lee Y, Kwon I, Jang Y, Song W, Cosio-Lima L and Roltsch M (2017) Potential signaling pathways of acute endurance exercise-induced cardiac autophagy and mitophagy and its possible role in cardioprotection, The Journal of Physiological Sciences, 10.1007/s12576-017-0555-7, 67:6, (639-654), Online publication date: 1-Nov-2017. Xue Q, Pei H, Liu Q, Zhao M, Sun J, Gao E, Ma X and Tao L (2017) MICU1 protects against myocardial ischemia/reperfusion injury and its control by the importer receptor Tom70, Cell Death & Disease, 10.1038/cddis.2017.280, 8:7, (e2923-e2923), Online publication date: 1-Jul-2017. He S, Wang X, Zhong Y, Tang L, Zhang Y, Ling Y, Tan Z, Yang P and Chen A (2017) Hesperetin post-treatment prevents rat cardiomyocytes from hypoxia/reoxygenation injury in vitro via activating PI3K/Akt signaling pathway, Biomedicine & Pharmacotherapy, 10.1016/j.biopha.2017.05.003, 91, (1106-1112), Online publication date: 1-Jul-2017. Pohjoismäki J and Goffart S (2017) The role of mitochondria in cardiac development and protection, Free Radical Biology and Medicine, 10.1016/j.freeradbiomed.2017.02.032, 106, (345-354), Online publication date: 1-May-2017. Li C, Zhang C, Wang T, Xuan J, Su C and Wang Y (2016) Heme oxygenase 1 induction protects myocardiac cells against hypoxia/reoxygenation-induced apoptosisHämoxygenase-1-Induktion schützt Myokardzellen gegen durch Hypoxie/Reoxygenierung induzierte Apoptose, Herz, 10.1007/s00059-016-4424-6, 41:8, (715-724), Online publication date: 1-Dec-2016. Yu J, Wu J, Xie P, Maimaitili Y, Wang J, Xia Z, Gao F, Zhang X and Zheng H (2016) Sevoflurane postconditioning attenuates cardiomyocyte hypoxia/reoxygenation injury via restoring mitochondrial morphology, PeerJ, 10.7717/peerj.2659, 4, (e2659) Li S, Liu C, Gu L, Wang L, Shang Y, Liu Q, Wan J, Shi J, Wang F, Xu Z, Ji G and Li W (2016) Autophagy protects cardiomyocytes from the myocardial ischaemia-reperfusion injury through the clearance of CLP36, Open Biology, 10.1098/rsob.160177, 6:8, (160177), Online publication date: 1-Aug-2016. Xia Y, Liu Y, Xia T, Li X, Huo C, Jia X, Wang L, Xu R, Wang N, Zhang M, Li H and Wang X (2016) Activation of volume-sensitive Cl− channel mediates autophagy-related cell death in myocardial ischaemia/reperfusion injury, Oncotarget, 10.18632/oncotarget.10050, 7:26, (39345-39362), Online publication date: 28-Jun-2016. Song Y, Shan J, Xue Z, Wang S, Xu H, Liu Y, Guo Y and Ren X (2016) Remote Postconditioning Induced by Trauma Protects the Mouse Heart against Ischemia Reperfusion Injury. Involvement of the Neural Pathway and Molecular Mechanisms, Cardiovascular Drugs and Therapy, 10.1007/s10557-016-6661-6, 30:3, (271-280), Online publication date: 1-Jun-2016. Zhang Y and Ren J (2016) Epigenetics and obesity cardiomyopathy: From pathophysiology to prevention and management, Pharmacology & Therapeutics, 10.1016/j.pharmthera.2016.03.005, 161, (52-66), Online publication date: 1-May-2016. Li Y, Wang S, Liu B, Tang L, Kuang R, Wang X, Zhao C, Song X, Cao X, Wu X, Yang P, Wang L and Chen A (2016) Sulforaphane prevents rat cardiomyocytes from hypoxia/reoxygenation injury in vitro via activating SIRT1 and subsequently inhibiting ER stress, Acta Pharmacologica Sinica, 10.1038/aps.2015.130, 37:3, (344-353), Online publication date: 1-Mar-2016. Ham O, Lee S, Lee C, Park J, Lee J, Seo H, Cha M, Choi E, Kim S and Hwang K (2015) let-7b suppresses apoptosis and autophagy of human mesenchymal stem cells transplanted into ischemia/reperfusion injured heart 7by targeting caspase-3, Stem Cell Research & Therapy, 10.1186/s13287-015-0134-x, 6:1, Online publication date: 1-Dec-2015. Gurianova V, Stroy D, Ciccocioppo R, Gasparova I, Petrovic D, Soucek M, Dosenko V and Kruzliak P (2015) Stress response factors as hub-regulators of microRNA biogenesis: implication to the diseased heart, Cell Biochemistry and Function, 10.1002/cbf.3151, 33:8, (509-518), Online publication date: 1-Dec-2015. Yu P, Zhang J, Yu S, Luo Z, Hua F, Yuan L, Zhou Z, Liu Q, Du X, Chen S, Zhang L, Xu G and Young M (2015) Protective Effect of Sevoflurane Postconditioning against Cardiac Ischemia/Reperfusion Injury via Ameliorating Mitochondrial Impairment, Oxidative Stress and Rescuing Autophagic Clearance, PLOS ONE, 10.1371/journal.pone.0134666, 10:8, (e0134666) WU D, JIANG H, CHEN S and ZHANG H (2015)(2015) Inhibition of microRNA-101 attenuates hypoxia/reoxygenation-induced apoptosis through induction of autophagy in H9c2 cardiomyocytes, Molecular Medicine Reports, 10.3892/mmr.2015.3215, 11:5, (3988-3994), Online publication date: 1-May-2015. Moresi V, Marroncelli N, Coletti D and Adamo S (2015) Regulation of skeletal muscle development and homeostasis by gene imprinting, histone acetylation and microRNA, Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, 10.1016/j.bbagrm.2015.01.002, 1849:3, (309-316), Online publication date: 1-Mar-2015. Xu J, Qin X, Cai X, Yang L, Xing Y, Li J, Zhang L, Tang Y, Liu J, Zhang X and Gao F (2015) Mitochondrial JNK activation triggers autophagy and apoptosis and aggravates myocardial injury following ischemia/reperfusion, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 10.1016/j.bbadis.2014.05.012, 1852:2, (262-270), Online publication date: 1-Feb-2015. Mei Y, Thompson M, Cohen R and Tong X (2015) Autophagy and oxidative stress in cardiovascular diseases, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 10.1016/j.bbadis.2014.05.005, 1852:2, (243-251), Online publication date: 1-Feb-2015. Zheng Q, Zhao K, Han X, Huff A, Cui Q, Babcock S, Yu S and Zhang Y (2015) Inhibition of AMPK accentuates prolonged caloric restriction-induced change in cardiac contractile function through disruption of compensatory autophagy, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 10.1016/j.bbadis.2014.04.023, 1852:2, (332-342), Online publication date: 1-Feb-2015. Zhang J, Wang C, Yu S, Luo Z, Chen Y, Liu Q, Hua F, Xu G and Yu P (2014) Sevoflurane Postconditioning Protects Rat Hearts against Ischemia-Reperfusion Injury via the Activation of PI3K/AKT/mTOR Signaling, Scientific Reports, 10.1038/srep07317, 4:1, Online publication date: 17-Dec-2014. Chen Z, Martin M, Li Z and Shyy J (2014) Endothelial dysfunction, Current Opinion in Lipidology, 10.1097/MOL.0000000000000107, 25:5, (339-349), Online publication date: 1-Oct-2014. March 11, 2014Vol 129, Issue 10 Advertisement Article InformationMetrics © 2014 American Heart Association, Inc.https://doi.org/10.1161/CIRCULATIONAHA.113.008115PMID: 24396040 Originally publishedJanuary 6, 2014 Keywordsautophagymyocardial ischemiaEditorialsPDF download Advertisement SubjectsAnimal Models of Human DiseaseCardiorenal SyndromeHeart Failure