Skip to main content
Top
Published in: Cardiovascular Drugs and Therapy 3/2010

01-06-2010

Mechanism of Cardioprotection by Early Ischemic Preconditioning

Authors: Xiulan Yang, Michael V. Cohen, James M. Downey

Published in: Cardiovascular Drugs and Therapy | Issue 3/2010

Login to get access

Abstract

A series of brief ischemia/reperfusion cycles (termed ischemic preconditioning, IPC) limits myocardial injury produced by a subsequent prolonged period of coronary artery occlusion and reperfusion. Over the last 2 decades our understanding of IPC’s mechanism has increased exponentially. Hearts exposed to IPC have a better metabolic and ionic status during prolonged ischemia compared to naïve hearts. However, this difference is not thought to be the main mechanism by which IPC protects against infarction. Signaling pathways that are activated by IPC distinguish IPC hearts from naïve hearts. During the trigger phase of IPC, adenosine, bradykinin and opioid receptors are occupied. Although these three receptors trigger signaling through divergent pathways, the signaling converges on protein kinase C. We have proposed that at the end of the index ischemia the activated PKC sensitizes the low-affinity A2b adenosine receptor (A2bAR) through phosphorylation of either the receptor or its coupling proteins so that A2bAR can be activated by endogenous adenosine released by the previously ischemic cardiomyocytes. The sensitized A2bAR would then be responsible for activation of the survival kinases including PI3 kinase, Akt and ERK which then act to inhibit lethal mitochondrial permeability transition pore formation which normally uncouples mitochondria and destroys many myocytes in the first minutes of reperfusion. Herein we review the evidence for the above mechanisms and their functional details.
Literature
1.
go back to reference Cohen MV, Downey JM. Myocardial preconditioning promises to be a novel approach to the treatment of ischemic heart disease. Annu Rev Med. 1996;47:21–9.PubMedCrossRef Cohen MV, Downey JM. Myocardial preconditioning promises to be a novel approach to the treatment of ischemic heart disease. Annu Rev Med. 1996;47:21–9.PubMedCrossRef
2.
go back to reference Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74:1124–36.PubMed Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74:1124–36.PubMed
3.
go back to reference Murry CE, Richard VJ, Jennings RB, Reimer KA. Myocardial protection is lost before contractile function recovers from ischemic preconditioning. Am J Physiol. 1991;260:H796–804.PubMed Murry CE, Richard VJ, Jennings RB, Reimer KA. Myocardial protection is lost before contractile function recovers from ischemic preconditioning. Am J Physiol. 1991;260:H796–804.PubMed
4.
go back to reference Jennings RB, Sebbag L, Schwartz LM, et al. Metabolism of preconditioned myocardium: effect of loss and reinstatement of cardioprotection. J Mol Cell Cardiol. 2001;33:1571–88.PubMedCrossRef Jennings RB, Sebbag L, Schwartz LM, et al. Metabolism of preconditioned myocardium: effect of loss and reinstatement of cardioprotection. J Mol Cell Cardiol. 2001;33:1571–88.PubMedCrossRef
5.
go back to reference Fleet WF, Johnson TA, Graebner CA, Gettes LS. Effect of serial brief ischemic episodes on extracellular K+, pH, and activation in the pig. Circulation. 1985;72:922–32.PubMed Fleet WF, Johnson TA, Graebner CA, Gettes LS. Effect of serial brief ischemic episodes on extracellular K+, pH, and activation in the pig. Circulation. 1985;72:922–32.PubMed
6.
go back to reference Murry CE, Richard VJ, Reimer KA, Jennings RB. Ischemic preconditioning slows energy metabolism and delays ultrastructural damage during a sustained ischemic episode. Circ Res. 1990;66:913–31.PubMed Murry CE, Richard VJ, Reimer KA, Jennings RB. Ischemic preconditioning slows energy metabolism and delays ultrastructural damage during a sustained ischemic episode. Circ Res. 1990;66:913–31.PubMed
7.
go back to reference Soares PR, de Albuquerque CP, Chacko VP, et al. Role of preischemic glycogen depletion in the improvement of postischemic metabolic and contractile recovery of ischemia-preconditioned rat hearts. Circulation. 1997;96:975–83.PubMed Soares PR, de Albuquerque CP, Chacko VP, et al. Role of preischemic glycogen depletion in the improvement of postischemic metabolic and contractile recovery of ischemia-preconditioned rat hearts. Circulation. 1997;96:975–83.PubMed
8.
go back to reference Miura T, Suzuki K, Shimamoto K, Iimura O. Suppression of the degradation of adenine nucleotides during ischemia may not be a sufficient mechanism for infarct size limitation by preconditioning. Basic Res Cardiol. 1996;91:425–32.PubMedCrossRef Miura T, Suzuki K, Shimamoto K, Iimura O. Suppression of the degradation of adenine nucleotides during ischemia may not be a sufficient mechanism for infarct size limitation by preconditioning. Basic Res Cardiol. 1996;91:425–32.PubMedCrossRef
9.
go back to reference Weinbrenner C, Wang P, Downey JM. Loss of glycogen during preconditioning is not a prerequisite for protection of the rabbit heart. Basic Res Cardiol. 1996;91:374–81.PubMed Weinbrenner C, Wang P, Downey JM. Loss of glycogen during preconditioning is not a prerequisite for protection of the rabbit heart. Basic Res Cardiol. 1996;91:374–81.PubMed
10.
go back to reference Yang X-M, Liu Y, Liu Y, et al. Attenuation of infarction in cynomolgus monkeys: preconditioning and postconditioning. Basic Res Cardiol. 2010;105:119–28.PubMedCrossRef Yang X-M, Liu Y, Liu Y, et al. Attenuation of infarction in cynomolgus monkeys: preconditioning and postconditioning. Basic Res Cardiol. 2010;105:119–28.PubMedCrossRef
11.
go back to reference Hausenloy DJ, Yellon DM. New directions for protecting the heart against ischaemia-reperfusion injury: targeting the Reperfusion Injury Salvage Kinase (RISK)-pathway. Cardiovasc Res. 2004;61:448–60.PubMedCrossRef Hausenloy DJ, Yellon DM. New directions for protecting the heart against ischaemia-reperfusion injury: targeting the Reperfusion Injury Salvage Kinase (RISK)-pathway. Cardiovasc Res. 2004;61:448–60.PubMedCrossRef
12.
go back to reference Solenkova NV, Solodushko V, Cohen MV, Downey JM. Endogenous adenosine protects preconditioned heart during early minutes of reperfusion by activating Akt. Am J Physiol. 2006;290:H441–9. Solenkova NV, Solodushko V, Cohen MV, Downey JM. Endogenous adenosine protects preconditioned heart during early minutes of reperfusion by activating Akt. Am J Physiol. 2006;290:H441–9.
13.
go back to reference Zhao Z-Q, Corvera JS, Halkos ME, et al. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. Am J Physiol. 2003;285:H579–88. Zhao Z-Q, Corvera JS, Halkos ME, et al. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. Am J Physiol. 2003;285:H579–88.
14.
go back to reference Förster K, Paul I, Solenkova N, et al. NECA at reperfusion limits infarction and inhibits formation of the mitochondrial permeability transition pore by activating p70S6 kinase. Basic Res Cardiol. 2006;101:319–26.PubMedCrossRef Förster K, Paul I, Solenkova N, et al. NECA at reperfusion limits infarction and inhibits formation of the mitochondrial permeability transition pore by activating p70S6 kinase. Basic Res Cardiol. 2006;101:319–26.PubMedCrossRef
15.
go back to reference Methner C, Donat U, Felix SB, Krieg T. Cardioprotection of bradykinin at reperfusion involves transactivation of the epidermal growth factor receptor via matrix metalloproteinase-8. Acta Physiol. 2009;197:265–71.CrossRef Methner C, Donat U, Felix SB, Krieg T. Cardioprotection of bradykinin at reperfusion involves transactivation of the epidermal growth factor receptor via matrix metalloproteinase-8. Acta Physiol. 2009;197:265–71.CrossRef
16.
go back to reference Liu GS, Thornton J, Van Winkle DM, et al. Protection against infarction afforded by preconditioning is mediated by A1 adenosine receptors in rabbit heart. Circulation. 1991;84:350–6.PubMed Liu GS, Thornton J, Van Winkle DM, et al. Protection against infarction afforded by preconditioning is mediated by A1 adenosine receptors in rabbit heart. Circulation. 1991;84:350–6.PubMed
17.
go back to reference Wall TM, Sheehy R, Hartman JC. Role of bradykinin in myocardial preconditioning. J Pharmacol Exp Ther. 1994;270:681–9.PubMed Wall TM, Sheehy R, Hartman JC. Role of bradykinin in myocardial preconditioning. J Pharmacol Exp Ther. 1994;270:681–9.PubMed
18.
go back to reference Schultz JEJ, Rose E, Yao Z, Gross GJ. Evidence for involvement of opioid receptors in ischemic preconditioning in rat hearts. Am J Physiol. 1995;268:H2157–61.PubMed Schultz JEJ, Rose E, Yao Z, Gross GJ. Evidence for involvement of opioid receptors in ischemic preconditioning in rat hearts. Am J Physiol. 1995;268:H2157–61.PubMed
19.
go back to reference Goto M, Liu Y, Yang X-M, et al. Role of bradykinin in protection of ischemic preconditioning in rabbit hearts. Circ Res. 1995;77:611–21.PubMed Goto M, Liu Y, Yang X-M, et al. Role of bradykinin in protection of ischemic preconditioning in rabbit hearts. Circ Res. 1995;77:611–21.PubMed
20.
go back to reference Cohen MV, Yang X-M, Liu GS, et al. Acetylcholine, bradykinin, opioids, and phenylephrine, but not adenosine, trigger preconditioning by generating free radicals and opening mitochondrial KATP channels. Circ Res. 2001;89:273–8.PubMedCrossRef Cohen MV, Yang X-M, Liu GS, et al. Acetylcholine, bradykinin, opioids, and phenylephrine, but not adenosine, trigger preconditioning by generating free radicals and opening mitochondrial KATP channels. Circ Res. 2001;89:273–8.PubMedCrossRef
21.
go back to reference Cohen MV, Philipp S, Krieg T, et al. Preconditioning-mimetics bradykinin and DADLE activate PI3-kinase through divergent pathways. J Mol Cell Cardiol. 2007;42:842–51.PubMedCrossRef Cohen MV, Philipp S, Krieg T, et al. Preconditioning-mimetics bradykinin and DADLE activate PI3-kinase through divergent pathways. J Mol Cell Cardiol. 2007;42:842–51.PubMedCrossRef
22.
go back to reference Oldenburg O, Qin Q, Krieg T, et al. Bradykinin induces mitochondrial ROS generation via NO, cGMP, PKG, and mitoKATP channel opening and leads to cardioprotection. Am J Physiol. 2004;286:H468–76. Oldenburg O, Qin Q, Krieg T, et al. Bradykinin induces mitochondrial ROS generation via NO, cGMP, PKG, and mitoKATP channel opening and leads to cardioprotection. Am J Physiol. 2004;286:H468–76.
23.
go back to reference Banerjee A, Locke-Winter C, Rogers KB, et al. Preconditioning against myocardial dysfunction after ischemia and reperfusion by an α1-adrenergic mechanism. Circ Res. 1993;73:656–70.PubMed Banerjee A, Locke-Winter C, Rogers KB, et al. Preconditioning against myocardial dysfunction after ischemia and reperfusion by an α1-adrenergic mechanism. Circ Res. 1993;73:656–70.PubMed
24.
go back to reference Liu Y, Tsuchida A, Cohen MV, Downey JM. Pretreatment with angiotensin II activates protein kinase C and limits myocardial infarction in isolated rabbit hearts. J Mol Cell Cardiol. 1995;27:883–92.PubMedCrossRef Liu Y, Tsuchida A, Cohen MV, Downey JM. Pretreatment with angiotensin II activates protein kinase C and limits myocardial infarction in isolated rabbit hearts. J Mol Cell Cardiol. 1995;27:883–92.PubMedCrossRef
25.
go back to reference Wang P, Gallagher KP, Downey JM, Cohen MV. Pretreatment with endothelin-1 mimics ischemic preconditioning against infarction in isolated rabbit heart. J Mol Cell Cardiol. 1996;28:579–88.PubMedCrossRef Wang P, Gallagher KP, Downey JM, Cohen MV. Pretreatment with endothelin-1 mimics ischemic preconditioning against infarction in isolated rabbit heart. J Mol Cell Cardiol. 1996;28:579–88.PubMedCrossRef
26.
go back to reference Kennedy S, Kane KA, Pyne NJ, Pyne S. Targeting sphingosine-1-phosphate signalling for cardioprotection. Curr Opin Pharmacol. 2009;9:194–201.PubMedCrossRef Kennedy S, Kane KA, Pyne NJ, Pyne S. Targeting sphingosine-1-phosphate signalling for cardioprotection. Curr Opin Pharmacol. 2009;9:194–201.PubMedCrossRef
27.
go back to reference Murry CE, Richard VJ, Jennings RB, Reimer KA. Preconditioning with ischemia: is the protective effect mediated by free radical-induced myocardial stunning? Circulation. 1988;78(Suppl II):II-77. Murry CE, Richard VJ, Jennings RB, Reimer KA. Preconditioning with ischemia: is the protective effect mediated by free radical-induced myocardial stunning? Circulation. 1988;78(Suppl II):II-77.
28.
go back to reference Baines CP, Goto M, Downey JM. Oxygen radicals released during ischemic preconditioning contribute to cardioprotection in the rabbit myocardium. J Mol Cell Cardiol. 1997;29:207–16.PubMedCrossRef Baines CP, Goto M, Downey JM. Oxygen radicals released during ischemic preconditioning contribute to cardioprotection in the rabbit myocardium. J Mol Cell Cardiol. 1997;29:207–16.PubMedCrossRef
29.
go back to reference Tritto I, D’Andrea D, Eramo N, et al. Oxygen radicals can induce preconditioning in rabbit hearts. Circ Res. 1997;80:743–8.PubMed Tritto I, D’Andrea D, Eramo N, et al. Oxygen radicals can induce preconditioning in rabbit hearts. Circ Res. 1997;80:743–8.PubMed
30.
go back to reference Vanden Hoek TL, Becker LB, Shao Z, et al. Reactive oxygen species released from mitochondria during brief hypoxia induce preconditioning in cardiomyocytes. J Biol Chem. 1998;273:18092–8.CrossRef Vanden Hoek TL, Becker LB, Shao Z, et al. Reactive oxygen species released from mitochondria during brief hypoxia induce preconditioning in cardiomyocytes. J Biol Chem. 1998;273:18092–8.CrossRef
31.
go back to reference Kuno A, Solenkova NV, Solodushko V, et al. Infarct limitation by a protein kinase G activator at reperfusion in rabbit hearts is dependent on sensitizing the heart to A2b agonists by protein kinase C. Am J Physiol. 2008;295:H1288–95. Kuno A, Solenkova NV, Solodushko V, et al. Infarct limitation by a protein kinase G activator at reperfusion in rabbit hearts is dependent on sensitizing the heart to A2b agonists by protein kinase C. Am J Physiol. 2008;295:H1288–95.
32.
go back to reference Liu Y, Yang X-M, Iliodromitis EK, et al. Redox signaling at reperfusion is required for protection from ischemic preconditioning but not from a direct PKC activator. Basic Res Cardiol. 2008;103:54–9.PubMedCrossRef Liu Y, Yang X-M, Iliodromitis EK, et al. Redox signaling at reperfusion is required for protection from ischemic preconditioning but not from a direct PKC activator. Basic Res Cardiol. 2008;103:54–9.PubMedCrossRef
33.
go back to reference Korichneva I, Hoyos B, Chua R, et al. Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen. J Biol Chem. 2002;277:44327–31.PubMedCrossRef Korichneva I, Hoyos B, Chua R, et al. Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen. J Biol Chem. 2002;277:44327–31.PubMedCrossRef
34.
go back to reference Kevin LG, Camara AKS, Riess ML, et al. Ischemic preconditioning alters real-time measure of O2 radicals in intact hearts with ischemia and reperfusion. Am J Physiol. 2003;284:H566–74. Kevin LG, Camara AKS, Riess ML, et al. Ischemic preconditioning alters real-time measure of O2 radicals in intact hearts with ischemia and reperfusion. Am J Physiol. 2003;284:H566–74.
35.
go back to reference Becker LB. New concepts in reactive oxygen species and cardiovascular reperfusion physiology. Cardiovasc Res. 2004;61:461–70.PubMedCrossRef Becker LB. New concepts in reactive oxygen species and cardiovascular reperfusion physiology. Cardiovasc Res. 2004;61:461–70.PubMedCrossRef
36.
go back to reference Carroll R, Gant VA, Yellon DM. Mitochondrial KATP channel opening protects a human atrial-derived cell line by a mechanism involving free radical generation. Cardiovasc Res. 2001;51:691–700.PubMedCrossRef Carroll R, Gant VA, Yellon DM. Mitochondrial KATP channel opening protects a human atrial-derived cell line by a mechanism involving free radical generation. Cardiovasc Res. 2001;51:691–700.PubMedCrossRef
37.
go back to reference Forbes RA, Steenbergen C, Murphy E. Diazoxide-induced cardioprotection requires signaling through a redox-sensitive mechanism. Circ Res. 2001;88:802–9.PubMedCrossRef Forbes RA, Steenbergen C, Murphy E. Diazoxide-induced cardioprotection requires signaling through a redox-sensitive mechanism. Circ Res. 2001;88:802–9.PubMedCrossRef
38.
go back to reference Costa ADT, Jakob R, Costa CL, et al. The mechanism by which the mitochondrial ATP-sensitive K+ channel opening and H2O2 inhibit the mitochondrial permeability transition. J Biol Chem. 2006;281:20801–8.PubMedCrossRef Costa ADT, Jakob R, Costa CL, et al. The mechanism by which the mitochondrial ATP-sensitive K+ channel opening and H2O2 inhibit the mitochondrial permeability transition. J Biol Chem. 2006;281:20801–8.PubMedCrossRef
39.
go back to reference Bolli R, Jeroudi MO, Patel BS, et al. Marked reduction of free radical generation and contractile dysfunction by antioxidant therapy begun at the time of reperfusion: evidence that myocardial “stunning” is a manifestation of reperfusion injury. Circ Res. 1989;65:607–22.PubMed Bolli R, Jeroudi MO, Patel BS, et al. Marked reduction of free radical generation and contractile dysfunction by antioxidant therapy begun at the time of reperfusion: evidence that myocardial “stunning” is a manifestation of reperfusion injury. Circ Res. 1989;65:607–22.PubMed
40.
go back to reference Costa ADT, Garlid KD, West IC, et al. Protein kinase G transmits the cardioprotective signal from cytosol to mitochondria. Circ Res. 2005;97:329–36.PubMedCrossRef Costa ADT, Garlid KD, West IC, et al. Protein kinase G transmits the cardioprotective signal from cytosol to mitochondria. Circ Res. 2005;97:329–36.PubMedCrossRef
41.
go back to reference Costa ADT, Garlid KD. Intramitochondrial signaling: interactions among mitoKATP, PKCε, ROS, and MPT. Am J Physiol. 2008;295:H874–82. Costa ADT, Garlid KD. Intramitochondrial signaling: interactions among mitoKATP, PKCε, ROS, and MPT. Am J Physiol. 2008;295:H874–82.
42.
go back to reference Oldenburg O, Qin Q, Sharma AR, et al. Acetylcholine leads to free radical production dependent on KATP channels, Gi proteins, phosphatidylinositol 3-kinase and tyrosine kinase. Cardiovasc Res. 2002;55:544–52.PubMedCrossRef Oldenburg O, Qin Q, Sharma AR, et al. Acetylcholine leads to free radical production dependent on KATP channels, Gi proteins, phosphatidylinositol 3-kinase and tyrosine kinase. Cardiovasc Res. 2002;55:544–52.PubMedCrossRef
43.
go back to reference Miro-Casas E, Ruiz-Meana M, Agullo E, et al. Connexin43 in cardiomyocyte mitochondria contributes to mitochondrial potassium uptake. Cardiovasc Res. 2009;83:747–56.PubMedCrossRef Miro-Casas E, Ruiz-Meana M, Agullo E, et al. Connexin43 in cardiomyocyte mitochondria contributes to mitochondrial potassium uptake. Cardiovasc Res. 2009;83:747–56.PubMedCrossRef
44.
go back to reference Heinzel FR, Luo Y, Li X, et al. Impairment of diazoxide-induced formation of reactive oxygen species and loss of cardioprotection in connexin 43 deficient mice. Circ Res. 2005;97:583–6.PubMedCrossRef Heinzel FR, Luo Y, Li X, et al. Impairment of diazoxide-induced formation of reactive oxygen species and loss of cardioprotection in connexin 43 deficient mice. Circ Res. 2005;97:583–6.PubMedCrossRef
45.
go back to reference Bell RM, Cave AC, Johar S, et al. Pivotal role of NOX-2-containing NADPH oxidase in early ischemic preconditioning. FASEB J. 2005;19:2037–9.PubMed Bell RM, Cave AC, Johar S, et al. Pivotal role of NOX-2-containing NADPH oxidase in early ischemic preconditioning. FASEB J. 2005;19:2037–9.PubMed
46.
go back to reference Ytrehus K, Liu Y, Downey JM. Preconditioning protects ischemic rabbit heart by protein kinase C activation. Am J Physiol. 1994;266:H1145–52.PubMed Ytrehus K, Liu Y, Downey JM. Preconditioning protects ischemic rabbit heart by protein kinase C activation. Am J Physiol. 1994;266:H1145–52.PubMed
47.
go back to reference Dorn GW 2nd, Souroujon MC, Liron T, et al. Sustained in vivo cardiac protection by a rationally designed peptide that causes ε protein kinase C translocation. PNAS. 1999;96:12798–803.PubMedCrossRef Dorn GW 2nd, Souroujon MC, Liron T, et al. Sustained in vivo cardiac protection by a rationally designed peptide that causes ε protein kinase C translocation. PNAS. 1999;96:12798–803.PubMedCrossRef
48.
go back to reference Ping P, Song C, Zhang J, et al. Formation of protein kinase Cε-Lck signaling modules confers cardioprotection. J Clin Invest. 2002;109:499–507.PubMed Ping P, Song C, Zhang J, et al. Formation of protein kinase Cε-Lck signaling modules confers cardioprotection. J Clin Invest. 2002;109:499–507.PubMed
49.
go back to reference Gray MO, Karliner JS, Mochly-Rosen D. A selective ε-protein kinase C antagonist inhibits protection of cardiac myocytes from hypoxia-induced cell death. J Biol Chem. 1997;272:30945–51.PubMedCrossRef Gray MO, Karliner JS, Mochly-Rosen D. A selective ε-protein kinase C antagonist inhibits protection of cardiac myocytes from hypoxia-induced cell death. J Biol Chem. 1997;272:30945–51.PubMedCrossRef
50.
go back to reference Liu GS, Cohen MV, Mochly-Rosen D, Downey JM. Protein kinase C-ε is responsible for the protection of preconditioning in rabbit cardiomyocytes. J Mol Cell Cardiol. 1999;31:1937–48.PubMedCrossRef Liu GS, Cohen MV, Mochly-Rosen D, Downey JM. Protein kinase C-ε is responsible for the protection of preconditioning in rabbit cardiomyocytes. J Mol Cell Cardiol. 1999;31:1937–48.PubMedCrossRef
51.
go back to reference Chen L, Hahn H, Wu G, et al. Opposing cardioprotective actions and parallel hypertrophic effects of δPKC and εPKC. Proc Natl Acad Sci. 2001;98:11114–9.PubMedCrossRef Chen L, Hahn H, Wu G, et al. Opposing cardioprotective actions and parallel hypertrophic effects of δPKC and εPKC. Proc Natl Acad Sci. 2001;98:11114–9.PubMedCrossRef
52.
go back to reference Inagaki K, Begley R, Ikeno F, Mochly-Rosen D. Cardioprotection by ε-protein kinase C activation from ischemia: continuous delivery and antiarrhythmic effect of an ε-protein kinase C-activating peptide. Circulation. 2005;111:44–50.PubMedCrossRef Inagaki K, Begley R, Ikeno F, Mochly-Rosen D. Cardioprotection by ε-protein kinase C activation from ischemia: continuous delivery and antiarrhythmic effect of an ε-protein kinase C-activating peptide. Circulation. 2005;111:44–50.PubMedCrossRef
53.
go back to reference Saurin AT, Pennington DJ, Raat NJH, et al. Targeted disruption of the protein kinase C epsilon gene abolishes the infarct size reduction that follows ischaemic preconditioning of isolated buffer-perfused mouse hearts. Cardiovasc Res. 2002;55:672–80.PubMedCrossRef Saurin AT, Pennington DJ, Raat NJH, et al. Targeted disruption of the protein kinase C epsilon gene abolishes the infarct size reduction that follows ischaemic preconditioning of isolated buffer-perfused mouse hearts. Cardiovasc Res. 2002;55:672–80.PubMedCrossRef
54.
go back to reference Hahn HS, Yussman MG, Toyokawa T, et al. Ischemic protection and myofibrillar cardiomyopathy: dose-dependent effects of in vivo δPKC inhibition. Circ Res. 2002;91:741–8.PubMedCrossRef Hahn HS, Yussman MG, Toyokawa T, et al. Ischemic protection and myofibrillar cardiomyopathy: dose-dependent effects of in vivo δPKC inhibition. Circ Res. 2002;91:741–8.PubMedCrossRef
55.
go back to reference Inagaki K, Hahn HS, Dorn GW 2nd, Mochly-Rosen D. Additive protection of the ischemic heart ex vivo by combined treatment with δ-protein kinase C inhibitor and ε-protein kinase C activator. Circulation. 2003;108:869–75.PubMedCrossRef Inagaki K, Hahn HS, Dorn GW 2nd, Mochly-Rosen D. Additive protection of the ischemic heart ex vivo by combined treatment with δ-protein kinase C inhibitor and ε-protein kinase C activator. Circulation. 2003;108:869–75.PubMedCrossRef
56.
go back to reference Kawamura S, Yoshida K-i, Miura T, et al. Ischemic preconditioning translocates PKC-δ and -ε, which mediate functional protection in isolated rat heart. Am J Physiol. 1998;275:H2266–71.PubMed Kawamura S, Yoshida K-i, Miura T, et al. Ischemic preconditioning translocates PKC-δ and -ε, which mediate functional protection in isolated rat heart. Am J Physiol. 1998;275:H2266–71.PubMed
57.
go back to reference Zhao J, Renner O, Wightman L, et al. The expression of constitutively active isotypes of protein kinase C to investigate preconditioning. J Biol Chem. 1998;273:23072–9.PubMedCrossRef Zhao J, Renner O, Wightman L, et al. The expression of constitutively active isotypes of protein kinase C to investigate preconditioning. J Biol Chem. 1998;273:23072–9.PubMedCrossRef
58.
go back to reference Mayr M, Metzler B, Chung Y-L, et al. Ischemic preconditioning exaggerates cardiac damage in PKC-δ null mice. Am J Physiol. 2004;287:H946–56. Mayr M, Metzler B, Chung Y-L, et al. Ischemic preconditioning exaggerates cardiac damage in PKC-δ null mice. Am J Physiol. 2004;287:H946–56.
59.
go back to reference Inagaki K, Chen L, Ikeno F, et al. Inhibition of δ-protein kinase C protects against reperfusion injury of the ischemic heart in vivo. Circulation. 2003;108:2304–7.PubMedCrossRef Inagaki K, Chen L, Ikeno F, et al. Inhibition of δ-protein kinase C protects against reperfusion injury of the ischemic heart in vivo. Circulation. 2003;108:2304–7.PubMedCrossRef
60.
go back to reference Inagaki K, Churchill E, Mochly-Rosen D. Epsilon protein kinase C as a potential therapeutic target for the ischemic heart. Cardiovasc Res. 2006;70:222–30.PubMedCrossRef Inagaki K, Churchill E, Mochly-Rosen D. Epsilon protein kinase C as a potential therapeutic target for the ischemic heart. Cardiovasc Res. 2006;70:222–30.PubMedCrossRef
61.
go back to reference Murphy E. Primary and secondary signaling pathways in early preconditioning that converge on the mitochondria to produce cardioprotection. Circ Res. 2004;94:7–16.PubMedCrossRef Murphy E. Primary and secondary signaling pathways in early preconditioning that converge on the mitochondria to produce cardioprotection. Circ Res. 2004;94:7–16.PubMedCrossRef
62.
go back to reference Kuno A, Critz SD, Cui L, et al. Protein kinase C protects preconditioned rabbit hearts by increasing sensitivity of adenosine A2b-dependent signaling during early reperfusion. J Mol Cell Cardiol. 2007;43:262–71.PubMedCrossRef Kuno A, Critz SD, Cui L, et al. Protein kinase C protects preconditioned rabbit hearts by increasing sensitivity of adenosine A2b-dependent signaling during early reperfusion. J Mol Cell Cardiol. 2007;43:262–71.PubMedCrossRef
63.
go back to reference Kitakaze M, Hori M, Takashima S, et al. Ischemic preconditioning increases adenosine release and 5′- nucleotidase activity during myocardial ischemia and reperfusion in dogs: implications for myocardial salvage. Circulation. 1993;87:208–15.PubMed Kitakaze M, Hori M, Takashima S, et al. Ischemic preconditioning increases adenosine release and 5′- nucleotidase activity during myocardial ischemia and reperfusion in dogs: implications for myocardial salvage. Circulation. 1993;87:208–15.PubMed
64.
go back to reference Kitakaze M, Hori M, Morioka T, et al. α1-Adrenoceptor activation increases ecto-5′-nucleotidase activity and adenosine release in rat cardiomyocytes by activating protein kinase C. Circulation. 1995;91:2226–34.PubMed Kitakaze M, Hori M, Morioka T, et al. α1-Adrenoceptor activation increases ecto-5′-nucleotidase activity and adenosine release in rat cardiomyocytes by activating protein kinase C. Circulation. 1995;91:2226–34.PubMed
65.
go back to reference Eckle T, Krahn T, Grenz A, et al. Cardioprotection by ecto-5′-nucleotidase (CD73) and A2B adenosine receptors. Circulation. 2007;115:1581–90.PubMedCrossRef Eckle T, Krahn T, Grenz A, et al. Cardioprotection by ecto-5′-nucleotidase (CD73) and A2B adenosine receptors. Circulation. 2007;115:1581–90.PubMedCrossRef
66.
go back to reference Eckle T, Köhler D, Lehmann R, et al. Hypoxia-inducible factor-1 is central to cardioprotection: a new paradigm for ischemic preconditioning. Circulation. 2008;118:166–75.PubMedCrossRef Eckle T, Köhler D, Lehmann R, et al. Hypoxia-inducible factor-1 is central to cardioprotection: a new paradigm for ischemic preconditioning. Circulation. 2008;118:166–75.PubMedCrossRef
67.
go back to reference Schulz R, Post H, Vahlhaus C, Heusch G. Ischemic preconditioning in pigs: a graded phenomenon. Its relation to adenosine and bradykinin. Circulation. 1998;98:1022–9.PubMed Schulz R, Post H, Vahlhaus C, Heusch G. Ischemic preconditioning in pigs: a graded phenomenon. Its relation to adenosine and bradykinin. Circulation. 1998;98:1022–9.PubMed
68.
go back to reference Goto M, Cohen MV, Van Wylen DGL, Downey JM. Attenuated purine production during subsequent ischemia in preconditioned rabbit myocardium is unrelated to the mechanism of protection. J Mol Cell Cardiol. 1996;28:447–54.PubMedCrossRef Goto M, Cohen MV, Van Wylen DGL, Downey JM. Attenuated purine production during subsequent ischemia in preconditioned rabbit myocardium is unrelated to the mechanism of protection. J Mol Cell Cardiol. 1996;28:447–54.PubMedCrossRef
69.
go back to reference Harrison GJ, Willis RJ, Headrick JP. Extracellular adenosine levels and cellular energy metabolism in ischemically preconditioned rat heart. Cardiovasc Res. 1998;40:74–87.PubMedCrossRef Harrison GJ, Willis RJ, Headrick JP. Extracellular adenosine levels and cellular energy metabolism in ischemically preconditioned rat heart. Cardiovasc Res. 1998;40:74–87.PubMedCrossRef
70.
go back to reference Philipp S, Yang X-M, Cui L, et al. Postconditioning protects rabbit hearts through a protein kinase C-adenosine A2b receptor cascade. Cardiovasc Res. 2006;70:308–14.PubMedCrossRef Philipp S, Yang X-M, Cui L, et al. Postconditioning protects rabbit hearts through a protein kinase C-adenosine A2b receptor cascade. Cardiovasc Res. 2006;70:308–14.PubMedCrossRef
71.
go back to reference Hausenloy DJ, Tsang A, Mocanu MM, Yellon DM. Ischemic preconditioning protects by activating prosurvival kinases at reperfusion. Am J Physiol. 2005;288:H971–6. Hausenloy DJ, Tsang A, Mocanu MM, Yellon DM. Ischemic preconditioning protects by activating prosurvival kinases at reperfusion. Am J Physiol. 2005;288:H971–6.
72.
go back to reference Baines CP, Wang L, Cohen MV, Downey JM. Myocardial protection by insulin is dependent on phosphatidylinositol 3-kinase but not protein kinase C or KATP channels in the isolated rabbit heart. Basic Res Cardiol. 1999;94:188–98.PubMedCrossRef Baines CP, Wang L, Cohen MV, Downey JM. Myocardial protection by insulin is dependent on phosphatidylinositol 3-kinase but not protein kinase C or KATP channels in the isolated rabbit heart. Basic Res Cardiol. 1999;94:188–98.PubMedCrossRef
73.
go back to reference Xu Z, Yang X-M, Cohen MV, et al. Limitation of infarct size in rabbit hearts by the novel adenosine receptor agonist AMP 579 administered at reperfusion. J Mol Cell Cardiol. 2000;32:2339–47.PubMedCrossRef Xu Z, Yang X-M, Cohen MV, et al. Limitation of infarct size in rabbit hearts by the novel adenosine receptor agonist AMP 579 administered at reperfusion. J Mol Cell Cardiol. 2000;32:2339–47.PubMedCrossRef
74.
go back to reference Albrecht B, Krahn T, Philipp S, et al. Selective adenosine A2b receptor activation mimics postconditioning in a rabbit infarct model. Circulation. 2006;114(Suppl II):II-14–5. Albrecht B, Krahn T, Philipp S, et al. Selective adenosine A2b receptor activation mimics postconditioning in a rabbit infarct model. Circulation. 2006;114(Suppl II):II-14–5.
75.
go back to reference Baxter GF, Mocanu MM, Brar BK, et al. Cardioprotective effects of transforming growth factor-β1 during early reoxygenation or reperfusion are mediated by p42/p44 MAPK. J Cardiovasc Pharmacol. 2001;38:930–9.PubMedCrossRef Baxter GF, Mocanu MM, Brar BK, et al. Cardioprotective effects of transforming growth factor-β1 during early reoxygenation or reperfusion are mediated by p42/p44 MAPK. J Cardiovasc Pharmacol. 2001;38:930–9.PubMedCrossRef
76.
go back to reference Schulman D, Latchman DS, Yellon DM. Urocortin protects the heart from reperfusion injury via upregulation of p42/p44 MAPK signaling pathway. Am J Physiol. 2002;283:H1481–8. Schulman D, Latchman DS, Yellon DM. Urocortin protects the heart from reperfusion injury via upregulation of p42/p44 MAPK signaling pathway. Am J Physiol. 2002;283:H1481–8.
77.
go back to reference Liao Z, Brar BK, Cai Q, et al. Cardiotrophin-1 (CT-1) can protect the adult heart from injury when added both prior to ischaemia and at reperfusion. Cardiovasc Res. 2002;53:902–10.PubMedCrossRef Liao Z, Brar BK, Cai Q, et al. Cardiotrophin-1 (CT-1) can protect the adult heart from injury when added both prior to ischaemia and at reperfusion. Cardiovasc Res. 2002;53:902–10.PubMedCrossRef
78.
go back to reference Yang X-M, Krieg T, Cui L, et al. NECA and bradykinin at reperfusion reduce infarction in rabbit hearts by signaling through PI3K, ERK, and NO. J Mol Cell Cardiol. 2004;36:411–21.PubMedCrossRef Yang X-M, Krieg T, Cui L, et al. NECA and bradykinin at reperfusion reduce infarction in rabbit hearts by signaling through PI3K, ERK, and NO. J Mol Cell Cardiol. 2004;36:411–21.PubMedCrossRef
79.
go back to reference Yang X-M, Philipp S, Downey JM, Cohen MV. Atrial natriuretic peptide administered just prior to reperfusion limits infarction in rabbit hearts. Basic Res Cardiol. 2006;101:311–8.PubMedCrossRef Yang X-M, Philipp S, Downey JM, Cohen MV. Atrial natriuretic peptide administered just prior to reperfusion limits infarction in rabbit hearts. Basic Res Cardiol. 2006;101:311–8.PubMedCrossRef
80.
go back to reference Cai Z, Semenza GL. Phosphatidylinositol-3-kinase signaling is required for erythropoietin-mediated acute protection against myocardial ischemia/reperfusion injury. Circulation. 2004;109:2050–3.PubMedCrossRef Cai Z, Semenza GL. Phosphatidylinositol-3-kinase signaling is required for erythropoietin-mediated acute protection against myocardial ischemia/reperfusion injury. Circulation. 2004;109:2050–3.PubMedCrossRef
81.
go back to reference Parsa CJ, Kim J, Riel RU, et al. Cardioprotective effects of erythropoietin in the reperfused ischemic heart: a potential role for cardiac fibroblasts. J Biol Chem. 2004;279:20655–62.PubMedCrossRef Parsa CJ, Kim J, Riel RU, et al. Cardioprotective effects of erythropoietin in the reperfused ischemic heart: a potential role for cardiac fibroblasts. J Biol Chem. 2004;279:20655–62.PubMedCrossRef
82.
go back to reference Hausenloy DJ, Ong S-B, Yellon DM. The mitochondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol. 2009;104:189–202.PubMedCrossRef Hausenloy DJ, Ong S-B, Yellon DM. The mitochondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol. 2009;104:189–202.PubMedCrossRef
83.
go back to reference Kitakaze M, Asakura M, Kim J, et al. Human atrial natriuretic peptide and nicorandil as adjuncts to reperfusion treatment for acute myocardial infarction (J-WIND): two randomised trials. Lancet. 2007;370:1483–93.PubMedCrossRef Kitakaze M, Asakura M, Kim J, et al. Human atrial natriuretic peptide and nicorandil as adjuncts to reperfusion treatment for acute myocardial infarction (J-WIND): two randomised trials. Lancet. 2007;370:1483–93.PubMedCrossRef
84.
go back to reference Piot C, Croisille P, Staat P, et al. Effect of cyclosporine on reperfusion injury in acute myocardial infarction. N Engl J Med. 2008;359:473–81.PubMedCrossRef Piot C, Croisille P, Staat P, et al. Effect of cyclosporine on reperfusion injury in acute myocardial infarction. N Engl J Med. 2008;359:473–81.PubMedCrossRef
85.
go back to reference Hunter DR, Haworth RA, Southard JH. Relationship between configuration, function, and permeability in calcium-treated mitochondria. J Biol Chem. 1976;251:5069–77.PubMed Hunter DR, Haworth RA, Southard JH. Relationship between configuration, function, and permeability in calcium-treated mitochondria. J Biol Chem. 1976;251:5069–77.PubMed
86.
go back to reference Crompton M, Costi A. Kinetic evidence for a heart mitochondrial pore activated by Ca2+, inorganic phosphate and oxidative stress. A potential mechanism for mitochondrial dysfunction during cellular Ca2+ overload. Eur J Biochem. 1988;178:489–501.PubMedCrossRef Crompton M, Costi A. Kinetic evidence for a heart mitochondrial pore activated by Ca2+, inorganic phosphate and oxidative stress. A potential mechanism for mitochondrial dysfunction during cellular Ca2+ overload. Eur J Biochem. 1988;178:489–501.PubMedCrossRef
87.
go back to reference Crompton M, Ellinger H, Costi A. Inhibition by cyclosporin A of a Ca2+-dependent pore in heart mitochondria activated by inorganic phosphate and oxidative stress. Biochem J. 1988;255:357–60.PubMed Crompton M, Ellinger H, Costi A. Inhibition by cyclosporin A of a Ca2+-dependent pore in heart mitochondria activated by inorganic phosphate and oxidative stress. Biochem J. 1988;255:357–60.PubMed
88.
go back to reference Nazareth W, Yafei N, Crompton M. Inhibition of anoxia-induced injury in heart myocytes by cyclosporin A. J Mol Cell Cardiol. 1991;23:1351–4.PubMedCrossRef Nazareth W, Yafei N, Crompton M. Inhibition of anoxia-induced injury in heart myocytes by cyclosporin A. J Mol Cell Cardiol. 1991;23:1351–4.PubMedCrossRef
89.
go back to reference Griffiths EJ, Halestrap AP. Protection by cyclosporin A of ischemia/reperfusion-induced damage in isolated rat hearts. J Mol Cell Cardiol. 1993;25:1461–9.PubMedCrossRef Griffiths EJ, Halestrap AP. Protection by cyclosporin A of ischemia/reperfusion-induced damage in isolated rat hearts. J Mol Cell Cardiol. 1993;25:1461–9.PubMedCrossRef
90.
go back to reference Griffiths EJ, Halestrap AP. Mitochondrial non-specific pores remain closed during cardiac ischaemia, but open upon reperfusion. Biochem J. 1995;307:93–8.PubMed Griffiths EJ, Halestrap AP. Mitochondrial non-specific pores remain closed during cardiac ischaemia, but open upon reperfusion. Biochem J. 1995;307:93–8.PubMed
91.
go back to reference Hausenloy DJ, Maddock HL, Baxter GF, Yellon DM. Inhibiting mitochondrial permeability transition pore opening: a new paradigm for myocardial preconditioning? Cardiovasc Res. 2002;55:534–43.PubMedCrossRef Hausenloy DJ, Maddock HL, Baxter GF, Yellon DM. Inhibiting mitochondrial permeability transition pore opening: a new paradigm for myocardial preconditioning? Cardiovasc Res. 2002;55:534–43.PubMedCrossRef
92.
go back to reference Baines CP. The mitochondrial permeability transition pore and ischemia-reperfusion injury. Basic Res Cardiol. 2009;104:181–8.PubMedCrossRef Baines CP. The mitochondrial permeability transition pore and ischemia-reperfusion injury. Basic Res Cardiol. 2009;104:181–8.PubMedCrossRef
93.
go back to reference Baines CP. The molecular composition of the mitochondrial permeability transition pore. J Mol Cell Cardiol. 2009;46:850–7.PubMedCrossRef Baines CP. The molecular composition of the mitochondrial permeability transition pore. J Mol Cell Cardiol. 2009;46:850–7.PubMedCrossRef
94.
go back to reference Tong H, Imahashi K, Steenbergen C, Murphy E. Phosphorylation of glycogen synthase kinase-3β during preconditioning through a phosphatidylinositol-3-kinase-dependent pathway is cardioprotective. Circ Res. 2002;90:377–9.PubMedCrossRef Tong H, Imahashi K, Steenbergen C, Murphy E. Phosphorylation of glycogen synthase kinase-3β during preconditioning through a phosphatidylinositol-3-kinase-dependent pathway is cardioprotective. Circ Res. 2002;90:377–9.PubMedCrossRef
95.
go back to reference Gross ER, Hsu AK, Gross GJ. Opioid-induced cardioprotection occurs via glycogen synthase kinase β inhibition during reperfusion in intact rat hearts. Circ Res. 2004;94:960–6.PubMedCrossRef Gross ER, Hsu AK, Gross GJ. Opioid-induced cardioprotection occurs via glycogen synthase kinase β inhibition during reperfusion in intact rat hearts. Circ Res. 2004;94:960–6.PubMedCrossRef
96.
go back to reference Nishihara M, Miura T, Miki T, et al. Erythropoietin affords additional cardioprotection to preconditioned hearts by enhanced phosphorylation of glycogen synthase kinase-3β. Am J Physiol. 2006;291:H748–55. Nishihara M, Miura T, Miki T, et al. Erythropoietin affords additional cardioprotection to preconditioned hearts by enhanced phosphorylation of glycogen synthase kinase-3β. Am J Physiol. 2006;291:H748–55.
97.
go back to reference Korzick DH, Kostyak JC, Hunter JC, Saupe KW. Local delivery of PKCε-activating peptide mimics ischemic preconditioning in aged hearts through GSK-3β but not F1-ATPase inactivation. Am J Physiol. 2007;293:H2056–63. Korzick DH, Kostyak JC, Hunter JC, Saupe KW. Local delivery of PKCε-activating peptide mimics ischemic preconditioning in aged hearts through GSK-3β but not F1-ATPase inactivation. Am J Physiol. 2007;293:H2056–63.
98.
go back to reference Das S, Wong R, Rajapakse N, et al. Glycogen synthase kinase 3 inhibition slows mitochondrial adenine nucleotide transport and regulates voltage-dependent anion channel phosphorylation. Circ Res. 2008;103:983–91.PubMedCrossRef Das S, Wong R, Rajapakse N, et al. Glycogen synthase kinase 3 inhibition slows mitochondrial adenine nucleotide transport and regulates voltage-dependent anion channel phosphorylation. Circ Res. 2008;103:983–91.PubMedCrossRef
99.
go back to reference Obame FN, Plin-Mercier C, Assaly R, et al. Cardioprotective effect of morphine and a blocker of glycogen synthase kinase 3β, SB216763 [3-(2, 4-dichlorophenyl)-4(1-methyl-1H-indol-3-yl)-1H-pyrrole-2, 5-dione], via inhibition of the mitochondrial permeability transition pore. J Pharmacol Exp Ther. 2008;326:252–8.PubMedCrossRef Obame FN, Plin-Mercier C, Assaly R, et al. Cardioprotective effect of morphine and a blocker of glycogen synthase kinase 3β, SB216763 [3-(2, 4-dichlorophenyl)-4(1-methyl-1H-indol-3-yl)-1H-pyrrole-2, 5-dione], via inhibition of the mitochondrial permeability transition pore. J Pharmacol Exp Ther. 2008;326:252–8.PubMedCrossRef
100.
go back to reference Juhaszova M, Zorov DB, Kim S-H, et al. Glycogen synthase kinase-3β mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 2004;113:1535–49.PubMed Juhaszova M, Zorov DB, Kim S-H, et al. Glycogen synthase kinase-3β mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 2004;113:1535–49.PubMed
101.
go back to reference Garlid KD, Costa ADT, Quinlan CL, et al. Cardioprotective signaling to mitochondria. J Mol Cell Cardiol. 2009;46:858–66.PubMedCrossRef Garlid KD, Costa ADT, Quinlan CL, et al. Cardioprotective signaling to mitochondria. J Mol Cell Cardiol. 2009;46:858–66.PubMedCrossRef
102.
go back to reference Gross ER, Hsu AK, Gross GJ. GSK3β inhibition and KATP channel opening mediate acute opioid-induced cardioprotection at reperfusion. Basic Res Cardiol. 2007;102:341–9.PubMedCrossRef Gross ER, Hsu AK, Gross GJ. GSK3β inhibition and KATP channel opening mediate acute opioid-induced cardioprotection at reperfusion. Basic Res Cardiol. 2007;102:341–9.PubMedCrossRef
103.
go back to reference Nishino Y, Webb IG, Davidson SM, et al. Glycogen synthase kinase-3 inactivation is not required for ischemic preconditioning or postconditioning in the mouse. Circ Res. 2008;103:307–14.PubMedCrossRef Nishino Y, Webb IG, Davidson SM, et al. Glycogen synthase kinase-3 inactivation is not required for ischemic preconditioning or postconditioning in the mouse. Circ Res. 2008;103:307–14.PubMedCrossRef
104.
go back to reference Skyschally A, van Caster P, Boengler K, et al. Ischemic postconditioning in pigs: no causal role for RISK activation. Circ Res. 2009;104:15–8.PubMedCrossRef Skyschally A, van Caster P, Boengler K, et al. Ischemic postconditioning in pigs: no causal role for RISK activation. Circ Res. 2009;104:15–8.PubMedCrossRef
105.
go back to reference Xi J, McIntosh R, Shen X, et al. Adenosine A2A and A2B receptors work in concert to induce a strong protection against reperfusion injury in rat hearts. J Mol Cell Cardiol. 2009;47:684–90.PubMedCrossRef Xi J, McIntosh R, Shen X, et al. Adenosine A2A and A2B receptors work in concert to induce a strong protection against reperfusion injury in rat hearts. J Mol Cell Cardiol. 2009;47:684–90.PubMedCrossRef
106.
go back to reference Vinten-Johansen J, Thourani VH, Ronson RS, et al. Broad-spectrum cardioprotection with adenosine. Ann Thorac Surg. 1999;68:1942–8.PubMedCrossRef Vinten-Johansen J, Thourani VH, Ronson RS, et al. Broad-spectrum cardioprotection with adenosine. Ann Thorac Surg. 1999;68:1942–8.PubMedCrossRef
107.
go back to reference Kin H, Zatta AJ, Lofye MT, et al. Postconditioning reduces infarct size via adenosine receptor activation by endogenous adenosine. Cardiovasc Res. 2005;67:124–33.PubMedCrossRef Kin H, Zatta AJ, Lofye MT, et al. Postconditioning reduces infarct size via adenosine receptor activation by endogenous adenosine. Cardiovasc Res. 2005;67:124–33.PubMedCrossRef
108.
go back to reference Gomez L, Li B, Mewton N, et al. Inhibition of mitochondrial permeability transition pore opening: translation to patients. Cardiovasc Res. 2009;83:226–33.PubMedCrossRef Gomez L, Li B, Mewton N, et al. Inhibition of mitochondrial permeability transition pore opening: translation to patients. Cardiovasc Res. 2009;83:226–33.PubMedCrossRef
Metadata
Title
Mechanism of Cardioprotection by Early Ischemic Preconditioning
Authors
Xiulan Yang
Michael V. Cohen
James M. Downey
Publication date
01-06-2010
Publisher
Springer US
Published in
Cardiovascular Drugs and Therapy / Issue 3/2010
Print ISSN: 0920-3206
Electronic ISSN: 1573-7241
DOI
https://doi.org/10.1007/s10557-010-6236-x

Other articles of this Issue 3/2010

Cardiovascular Drugs and Therapy 3/2010 Go to the issue