Forum Minireview. Tetsuji Miura 1, *, Masahiro Nishihara 1, and Takayuki Miki 1

Similar documents
Reperfusion Injury: How Can We Reduce It?

Sean Davidson. The Hatter Cardiovascular Institute University College London, UK

Ischemia and Reperfusion: Pharmacological treatment options

Reactive oxygen species: Importance for ischemia/reperfusion (injury)

Postconditioning: from experimental proof to clinical concept

Ameliorating Reperfusion Injury During Resuscitation from Cardiac Arrest

NIH Public Access Author Manuscript Circ Res. Author manuscript; available in PMC 2009 October 24.

University of Milan spin-off founded in 2004 and now fully owned by Genextra S.p.A.

Glycogen Synthase Kinase 3 Inhibition Slows Mitochondrial Adenine Nucleotide Transport and Regulates Voltage-Dependent Anion Channel Phosphorylation

cardiac pre and post conditioning

Master Eudipharm 2012 Introductory Module, Principles of Discovery of Medicine and Development Planning. Nathan Mewton, MD, PhD. September 26 th 2012

Cardioprotezione ed invecchiamento

Vascular Biology. Rapid Communication

Ischemic preconditioning is cardioprotective 1 and depends

Pathophysiology of ischemia-reperfusion injury (and how to protect against it )

Reperfusion Effects After Cardiac Ischemia

Cell Injury MECHANISMS OF CELL INJURY

Tissue reperfusion after ischemia, although ultimately

Blockade of electron transport during ischemia preserves bcl-2 and inhibits opening of the mitochondrial permeability transition pore

The Role of the Mitochondrial Permeability Transition. Pore in Cardioprotection. Cara Hendry. Thesis submitted for the degree of MD (Research)

Ischemic Postconditioning During Primary Percutaneous Coronary Intervention Mechanisms and Clinical Application Jian Liu, MD FACC FESC FSCAI Chief Phy

EXPERIMENTAL AND THERAPEUTIC MEDICINE 8: , 2014

6. Lochner, A., Tromp, E., Mouton, R. Signal transduction in myocardial ischaemia and reperfusion. Mol Cell Biochem., 160/161: , 1996.

Jefferson Digital Commons

Prevention of reperfusion injury in STEMI - Contra

Reperfusion injury in STEMI. Therapeutic opportunities David Garcia-Dorado. Barcelona. Spain

There are numerous reviews on the signal transduction of

Sevoflurane confers additive cardioprotection to ethanol preconditioning associated with enhanced

Mechanisms of Cell Injury

CPR flow to prime the ischemic heart during cardiac arrest?

Glycogen Synthase Kinase-3 Inactivation Is Not Required for Ischemic Preconditioning or Postconditioning in the Mouse

Adjunctive Therapy to Reduce Infarct Size: Current and Future Challenges

Effect of Cyclosporine on Left Ventricular Remodeling After Reperfused Myocardial Infarction

Angiotensin-converting enzyme inhibitors potentiate subthreshold preconditioning through NO and mitok ATP. channel

Joost Seeger. A thesis submitted in partial fulfilment of the requirements of Liverpool John Moores University for the degree of Doctor of Philosophy.

Pathophysiology and treatment of focal cerebral ischemia

Nuovi target e opportunità terapeutiche del danno da riperfusione nello STEMI

Myocardial injury that has developed through a

REVIEWbph_ BJP. Introduction. British Journal of Pharmacology. WY Lim 1, CM Messow 2 and C Berry 1

Supplementary Figure 1. Confocal immunofluorescence showing mitochondrial translocation of Drp1. Cardiomyocytes treated with H 2 O 2 were prestained

Ginkgo biloba extract postconditioning reduces myocardial ischemia reperfusion injury

AperTO - Archivio Istituzionale Open Access dell'università di Torino

Metabolism of cardiac muscle. Dr. Mamoun Ahram Cardiovascular system, 2013

Reduced Susceptibility to Ischemia-Induced Arrhythmias in the Preconditioned Rat Heart Is Independent of PI3-Kinase/Akt

Imaging ischemic strokes: Correlating radiological findings with the pathophysiological evolution of an infarct

Cyclosporin A and cardioprotection: from investigative tool to therapeutic agent

Berbamine Protects the Heart From Ischemia/Reperfusion Injury by Maintaining Cytosolic Ca 2+ Homeostasis and Preventing Calpain Activation

S. Lemoine 1, *, L. Tritapepe 2, J. L. Hanouz 1 and P. E. Puddu 2. Abstract REVIEW ARTICLES

Chapter 14 - Electron Transport and Oxidative Phosphorylation

AT1 RECEPTOR BLOCKADE ATTENUATES INSULIN RESISTANCE AND MYOCARDIAL REMODELING IN RATS WITH DIET-INDUCED OBESITY

Effect of mitochondrial ATP-sensitive potassium channel opening on the translocation of protein kinase C epsilon in adult rat ventricular myocytes

Remote Ischemic Preconditioning: Current aspects of mechanisms. Hans Erik Bøtker, Aarhus University Hospital, Skejby, Denmark,

Cell therapy: enhancing the therapeutic potential of cardiac progenitors for delivery post myocardial infarction. Rita Alonaizan

Cell Signaling part 2

PUBLICATIONS. cells. J. Physiol. (London) 517P:91P (Manchester, England, UK).

Novel Necrosis Inhibitor to prevent. Myocardial Ischemia-Reperfusion Injury

BIOCHEMICAL EXAMINATION OF ACUTE MYOCARDIAL INFARCTION. Written by Lenka Fialová, translated by Jan Pláteník

c Ischemia (30 min) Reperfusion (8 w) Supplementary Figure bp 300 bp Ischemia (30 min) Reperfusion (4 h) Dox 20 mg/kg i.p.

Introduction ORIGINAL RESEARCH. Helen E. Turrell, Chokanan Thaitirarot, Hayley Crumbie & Glenn Rodrigo. Abstract

Original Article Change of mitochondrial function in the early stage after cardiac ischemia-reperfusion injury in mice

Thyroid hormone and cardiac regeneration: from Prometheus myth to reality

Medical Biochemistry and Molecular Biology department

BEER AND THE HEART: INSIGHTS ON THE EFFECTS ON ALCOHOLIC AND NON-ALCOHOLIC BEER

Crosstalk between Adiponectin and IGF-IR in breast cancer. Prof. Young Jin Suh Department of Surgery The Catholic University of Korea

Molecular Basis of Cardioprotection by Erythropoietin

Correlations among copeptin, ischemia-modified albumin, and the extent of myocardial injury in patients with acute carbon monoxide poisoning

ELECTROPHYSIOLOGICAL CHANGES DURING MYOCARDIAL ISCHAEMIA

Ischaemic Preconditioning prevents the differentiation induced by ischaemia/reperfusion injury of rat cardiac fibroblast to myofibroblast

ETIOLOGY AND PATHOGENESIS OF HYPOXIC-ISCHEMIC ENCEPHALOPATHY

Growth and Differentiation Phosphorylation Sampler Kit

Exercise in Adverse Cardiac Remodeling: of Mice and Men

Defects in glucose utilization & GLP-1

Receptor mediated Signal Transduction

Reperfusion Injury, Cardioprotection, and 2 Decades of Failed Studies

Mechanistic Toxicology

Mechanisms involved in the desflurane-induced post-conditioning of isolated human right atria from patients with type 2 diabetes

Phospho-AKT Sampler Kit

Interaction of Cardiovascular Risk Factors with Myocardial Ischemia/Reperfusion Injury, Preconditioning, and Postconditioning

Journal of the American College of Cardiology Vol. 38, No. 4, by the American College of Cardiology ISSN /01/$20.

Equivalent Cardioprotection Induced by Ischemic and Hypoxic Preconditioning

ATEF ELBAHRY,FACA,FICA,MISCP,FVBWG.

STEMI: Evolving Early Therapies of Myocardial Ischemia/Reperfusion Injury.

Pathophysiology and treatment of focal cerebral ischemia

Protein kinase A mediated stimulation of activating transcription factor 3 by hypertrophic stimuli in cardiomyocytes

Difficult Scenarios for Myocardial Protection SAHA Gil Bolotin M.D., Ph.D. Rambam Medical Center, Haifa, Israel

Chemical and Biochemical Mechanism Of Cell Injury.

Myocardial postconditioning: anesthetic considerations

Reactive Oxygen Species Induced Stimulation of 5 AMP-Activated Protein Kinase Mediates Sevoflurane-Induced Cardioprotection

Part-4. Cell cycle regulatory protein 5 (Cdk5) A novel target of ERK in Carb induced cell death

NIH Public Access Author Manuscript Future Cardiol. Author manuscript; available in PMC 2013 September 01.

In the name of GOD. Animal models of cardiovascular diseases: myocardial infarction & hypertension

Role of protein kinase C-«(PKC«) in isoflurane-induced cardioprotection

KEY CONCEPT QUESTIONS IN SIGNAL TRANSDUCTION

A particular set of insults induces apoptosis (part 1), which, if inhibited, can switch to autophagy. At least in some cellular settings, autophagy se

Cardiovascular Research Advance Access published August 12, New pathophysiological function of protein phosphatase 2A?

Myocardial ischaemia reperfusion injury: the challenge of translating ischaemic and anaesthetic protection from animal models to humans

(This is a sample cover image for this issue. The actual cover is not yet available at this time.)

Atrial Natriuretic Peptide Protects Against Ischemia-Reperfusion Injury in the Isolated Rat Heart

Impact factor: Reporter:4A1H0019 Chen Zi Hao 4A1H0023 Huang Wan ting 4A1H0039 Sue Yi Zhu 4A1H0070 Lin Guan cheng 4A1H0077 Chen Bo xuan

Perioperative Management of TAPVC

Transcription:

J Pharmacol Sci 109, 162 167 (2009)2 Journal of Pharmacological Sciences 2009 The Japanese Pharmacological Society Forum Minireview Drug Development Targeting the Glycogen Synthase Kinase-3β (GSK-3β)- Mediated Signal Transduction Pathway: Role of GSK-3β in Myocardial Protection Against Ischemia/Reperfusion Injury Tetsuji Miura 1, *, Masahiro Nishihara 1, and Takayuki Miki 1 1 Second Department of Internal Medicine, Sapporo Medical University, School of Medicine, South-1 West-16, Chuo-ku, Sapporo 060-8543, Japan Received October 16, 2008; Accepted December 1, 2008 Abstract. Although reperfusion is required to salvage ischemic myocardium from necrosis, reperfusion per se induces myocardial necrosis. In this lethal reperfusion injury, opening of the mitochondrial permeability transition pore (mptp) upon reperfusion is crucially involved. The mptp primarily consists of adenine nucleotide translocator (ANT) and voltage-dependent anion channel, and its opening is triggered by binding of cyclophilin-d (CyP-D) to ANT, which increases Ca 2+ sensitivity of the mptp. Recent studies have shown that inactivation of glycogen synthase kinase-3β (GSK-3β) suppresses mptp opening and protects cardiomyocytes. Multiple intracellular signals relevant to cardiomyocyte protection converge to GSK-3β and inactivate this kinase by phosphorylation. Although the effect of GSK-3β phosphorylation on mptp structure and function remains unclear, suppression of ANT CyP-D interaction by binding of phospho- GSK-3β to ANT and reduction in GSK-3β mediated phosphorylation of p53 may contribute to elevation of the threshold for mptp opening. Furthermore, a significant inverse correlation was observed between level of phospho-gsk-3β at the time of reperfusion and the extent of myocardium infarction in heart. Together with the infarct size limiting effect of GSK-3β inhibitors, this finding indicates that phospho-gsk-3β is a determinant of myocardial tolerance against reperfusion-induced necrosis. Thus, GSK-3β appears to be a target of novel therapy for cardioprotection upon reperfusion. Keywords: glycogen synthase kinase-3β (GSK-3β), myocardial infarction, signal transduction, mitochondria, mitochondrial permeability transition pore Introduction Glycogen synthase kinase-3β (GSK-3β) is a constitutively active Ser/Thr protein kinase, the function of which is regulated by phosphorylation, intracellular translocation, and complex formation with other proteins (1, 2). Roles of GSK-3β in cardiac glycogen metabolism and its function in counteracting ventricular hypertrophy have been well characterized (3, 4). Recently, accumulating evidence suggests that GSK-3β is also critically involved in the fate of cells subjected to extracellular *Corresponding author. miura@sapmed.ac.jp Published online in J-STAGE on January 29, 2009 (in advance) doi: 10.1254/jphs.08R27FM stress, including ischemia/reperfusion (5 10). In this article, we review the current understanding of mechanisms of ischemia/reperfusion injury in cardiomyocytes and roles of GSK-3β in myocardial protection. Mechanisms underlying cardiomyocyte necrosis after ischemia/reperfusion After the coronary artery has been occluded, ischemic myocytes lose high-energy phosphates and starve to death unless blood flow is restored. However, salvage of ischemic myocardium by restoration of blood flow is compromised by reperfusion-induced injury. Recent animal studies (9, 11 16) have shown that pharmacological intervention (for example, administration of a 162

GSK-3β in Cardioprotection 163 Na + -Ca 2+ exchange inhibitor or an adenosine A 2b receptor agonist) or non-pharmacological intervention (for example, ischemic postconditioning) at the time of reperfusion can reduce infarct size from 50% of the ischemic zone to approximately 30%. Thus, it is clear that a substantial mass of myocardium that could have contributed to functional recovery after reperfusion is lost by lethal reperfusion injury. Reperfusion injury is a result of priming the myocardium with ischemic injury, which results in intracellular acidosis, Na + overload (by Na + -H + exchanger, un-inactivated Na + channels, and inhibited Na + -K + - ATPase), depletion of ATP (mainly by mitochondrial ATPase), mitochondrial Ca 2+ overload, and osmotic loading (11, 12, 17). Ca 2+ influx via Na + -Ca 2+ exchange is inhibited by acidosis after sustained ischemia, but abrupt reperfusion washes out extracellular protons and provides oxygen to injured mitochondria. As a result, rapid intracellular Ca 2+ overload caused by reversemode Na + -Ca 2+ exchange, burst production of reactive oxygen species (ROS) from mitochondria, marked cell edema, and hypercontracture of myocytes occur shortly after reperfusion (11, 12, 17, 18). Intracellular Ca 2+ overload both activates Ca 2+ -activated proteases, including calpain (19), and augments mitochondrial Ca 2+ overload. ROS induce oxidation of lipids and proteins, and hypercontracture of myocytes induces disruption of fragile sarcolemma. All of these reperfusion-triggered changes are detrimental to cardiomyocytes and contribute to cell necrosis. However, the fate of cardiomyocytes after reperfusion appears to heavily depend on mitochondrial function. Capacity of mitochondrial ATP production after reperfusion is crucial to quickly restore intracellular Na + level by Na + -K + ATPase, which is necessary for prevention of catastrophic Ca 2+ overload and rigor contracture (11, 12). An important mechanism of irreversible impairment of mitochondrial function after ischemia /reperfusion injury is opening of the mitochondrial permeability transition pore (mptp). mptp and cardiomyocyte necrosis The molecular structure of the mptp has not yet been unequivocally determined. However, in a current model, the mptp basically consists of a voltage-dependent anion channel (VDAC) in the outer membrane and adenine nucleotide translocator (ANT) in the inner membrane (20, 21). The mptp is closed under physiological conditions but opens in response to cellular stress, allowing passage of substances less than 1,500 Da. Unless it is transient and reversible, opening of the mptp abolishes transmembrane potential in the mitochondria and thus ATP generation. In experimental preparations, opening of the mptp has been assessed by uptake of radioactive 2-deoxyglucose (2-DG) into mitochondria, leakage of calcein trapped in mitochondria, loss of mitochondrial membrane potential, or irreversible Ca 2+ release from isolated mitochondria after repetitive loading of Ca 2+ (22 26). All of these techniques indicate that the mptp in the cardiomyocyte opens after ischemia/ reperfusion or hypoxia/ reoxygenation. Timing of mptp opening was found to be within 3 10 min after reperfusion, but not during ischemia, in buffer-perfused rat hearts (20, 22). Several factors have been shown to contribute to opening of the mptp (20, 21). The most important factor is mitochondrial Ca 2+, and loading of a large amount of Ca 2+ to isolated mitochondria alone can induce opening of the mptp. Depletion of ATP, elevation of inorganic phosphate level, and ROS are also known to facilitate opening of the mptp. However, an important trigger of mptp opening is a matrix protein, cyclophilin-d (CyP- D). Binding of this protein to ANT increases Ca 2+ sensitivity of ANT and thus reduces the threshold for opening of the mptp. Treatments with inhibitors of CyP-D (cyclosporine A and sangliferin A) and deletion of Ppif, a gene coding for mitochondrial CyP-D, significantly increased mitochondrial resistance to mptp opening in response to Ca 2+ overload and oxidant stress, indicating that CyP-D plays a major role in mptp opening (27 32). Contribution of CyP-D mediated opening of the mptp to myocardial necrosis after ischemia/reperfusion has been indicated by results of infarct size experiments. Infarct size was significantly reduced by pharmacological inhibitors of CyP-D and by knock-out of the mitochondrial CyP-D gene (20, 31, 33). Furthermore, cyclosporine A did not afford further infarct size limitation in CyP-D knock-out mice (33). GSK-3β as a regulator of mptp opening threshold GSK-3β has recently received attention as a possible regulator of mptp opening since this kinase is a common target of multiple signal pathways that lead to myocardial protection from infarction. In fact, myocardial infarct size is reduced by ischemic preconditioning and by treatment with δ-opioid, insulin, insulin-like growth factor, erythropoietin, or isoflurane; and all of these interventions induce Ser 9 -phosphorylation of GSK-3β (6 13, 16, 34, 35). However, different protein kinases (i.e., protein kinase C [PKC], Akt, ERK1/2) are responsible, depending on the intervention, for direct phosphorylation of GSK-3β in the cardioprotection. Evidence for a regulatory role of GSK-3β in mptp opening was first reported by Juhaszova et al. (5). They

164 T Miura et al determined the threshold for opening of the mptp by monitoring mitochondrial membrane potential in isolated cardiomyocytes and used ROS generated by laser irradiation as a trigger of mptp opening. The threshold for mptp opening was significantly elevated by inhibitory phosphorylation of GSK-3β at Ser 9, transfection of a dominant negative mutant of GSK-3β, or reduction of GSK-3β expression by sirna. Recently Gomez et al. (8) assessed the involvement of GSK-3β in inhibition of mptp opening by ischemic postconditioning. They isolated mitochondria from the myocardium after ischemia/reperfusion and determined the threshold for mptp opening as the amount of loading Ca 2+ required to induce irreversible Ca 2+ release from the mitochondria. Postconditioning significantly elevated the threshold of mptp opening in cardiac mitochondria from wild-type mice, but such a protective effect was not detected in mitochondria from transgenic mice expressing GSK- S9A, which cannot be inactivated by phosphorylation at Ser 9. Taken together, these findings support the notion that phospho-gsk-3β regulates the threshold for mptp opening in response to ROS and/or Ca 2+ overloading. To get an insight into the molecular mechanism by which phospho-gsk-3β regulates mptp opening, we examined intracellular localization of GSK-3β and interaction of GSK-3β with mptp before and after ischemia/ reperfusion in isolated perfused rat hearts (10). GSK-3β was predominantly found in the cytosolic fraction under baseline conditions, but ischemia/reperfusion significantly increased GSK-3β level in the mitochondrial fraction. Reperfusion also increased phospho-gsk-3β level in all cell fractions. GSK-3β was co-immunoprecipitated with ANT and with VDAC before ischemia, but levels of these complexes were increased after reperfusion by approximately 50%, indicating that GSK- 3β translocated after reperfusion binds to mptp. Based on the finding of reperfusion-induced GSK-3β interaction with ANT and VDAC, we hypothesized that elevation of the threshold for mptp opening by phosphorylation of GSK-3β is achieved by direct interaction of phospho-gsk-3β with mptp subunit proteins (10). To test this hypothesis, GSK-3β phosphorylation at Ser 9 was induced by ischemic preconditioning and erythropoietin-receptor activation in rat hearts. Phospho- GSK-3β in the reperfused myocardium was increased by preconditioning and erythropoietin-receptor activation in a PKC- and Akt-dependent manner, and the increased phospho-gsk-3β was co-immunoprecipitated with ANT but not with VDAC. Furthermore, the level of CyP-D co-immunoprecipitated with ANT was significantly reduced to 40% in association with a 50% increase in phospho-gsk-3β ANT binding (10). These results suggest that interaction of phospho-gsk-3β with ANT inhibits CyP-D ANT interaction, resulting in prevention of mptp opening. In addition to ANT, p53 may be involved in suppression of mptp opening by phosphorylation of GSK-3β. p53 is one of more than 20 substrates of GSK-3β, and its phosphorylation enhances functional activity and translocation of p53 to the nucleus and mitochondria (36). A recent study by Venkatapuram et al. (37) showed that an inhibitor of p53, pifithrin-α, lowered the threshold of isoflurane-induced limitation of infarct size in rabbit hearts. Interestingly, this beneficial effect of a p53 inhibitor was abolished by an activator of mptp, atractyloside. Thus, inhibition of GSK-3β mediated p53 phosphorylation may also contribute to suppression of myocardial necrosis by GSK-3β phosphorylation. Impairment of cell signaling upstream of GSK-3β by co-existing diseases There are redundant signal pathways that induce phosphorylation of GSK-3β in cardiomyocytes (6), and more than one pathway is frequently activated by cardioprotective receptor agonists and ischemic preconditioning (9, 16). However, some of the cytoprotective signal pathways suffer from impairment by concurrent diseases associated with coronary artery diseases (such as diabetes mellitus, hypercholesterolemia, and post-infarct ventricular remodeling) (38 42). We have found that post-infarct remodeling impairs activation of PKC-ε after ischemic preconditioning and Jak2 PI3K Akt signaling by erythropoietin-receptor activation (39 41). Erythropoietin-receptor activation also failed to activate the Jak2 PI3K Akt pathway in the myocardium of an animal model of type 2 diabetes (42). Thus, from the viewpoint of clinical application, a strategy to directly inhibit GSK-3β would be more preferable to indirect GSK-3β inhibition by use of its up-stream signaling pathways. Level of phospho-gsk-3β as a determinant of infarct size in hearts in situ Physiological parameters that determine the extent of myocardial necrosis (i.e., infarct size) after ischemia /reperfusion have been fully characterized, and duration of ischemia, size of the ischemic region, residual blood flow in the ischemic region, and myocardial oxygen consumption are major determinants of infarct size in hearts in situ (43, 44). However, a molecule that ultimately determines the level of tolerance of the cardiomyocyte against necrosis at the time of reperfusion has not been identified. GSK-3β is actually a candidate for such a molecule since multiple cytoprotec-

GSK-3β in Cardioprotection 165 tive signal pathways converge to this protein kinase (6, 9, 16) as discussed above. Using a rat model of myocardial infarction, we examined the possibility that level of phospho-gsk-3β at the time of reperfusion, when the mptp opens, is a determinant of infarct size in hearts in situ (9). To prepare different levels of GSK-3β phosphorylation by activating different signal pathways, we employed ischemic preconditioning, erythropoietin pretreatment, their combination, and pretreatments with protein kinase inhibitors. Infarct size after 20-min coronary artery occlusion/reperfusion in rat hearts in situ was significantly reduced by ischemic preconditioning and by erythropoietin pretreatment, and their combination afforded additive limitation of infarct size. Such an additive effect of ischemic preconditioning and erythropoietin pretreatment was observed for GSK-3β phosphorylation upon reperfusion but not for Akt phosphorylation or STAT3 phosphorylation. A PKC inhibitor (chelerythrine) and a PI3K inhibitor (wortmannin) partly inhibited both GSK-3β phosphorylation and infarct size limitation by preconditioning and erythropoietin. Interestingly, there was a tight inverse relationship between phospho-gsk-3β level upon reperfusion and infarct size 2 h after reperfusion (9). Furthermore, administration of an inhibitor of GSK-3β before reperfusion significantly limited infarct size as previously reported (6, 7). Taken together, these findings suggest that the level of phospho-gsk-3β at the time of reperfusion in the myocardium is indeed a determinant of infarct size in situ. Is a GSK-3β inhibitor a promising infarct size-limiting agent for clinical application? Introduction of reperfusion therapy has markedly improved prognosis of patients with acute myocardial infarction (AMI). However, current reperfusion therapy cannot afford sufficient myocardial salvage in approximately 25% of patients, who subsequently develop severe heart failure (45). Thus, there is a clinical need for novel therapy to protect cardiomyocytes from ischemia/ reperfusion-induced necrosis. An advantage of a GSK-3β inhibitor for clinical use is its efficiency even when administered immediately before reperfusion. Another favorable profile of GSK-3β inhibitors is that their protective effect would not be impaired by common co-morbidities in AMI patients that modify signaling to cytoprotective kinases such as PKC-ε and Akt (10, 41, 42, 46). Although chronic inhibition of GSK-3β raises concern about risks of cancer development and ventricular hypertrophy (1, 4), such risks would be negligible for single injection of a GSK-3β inhibitor before reperfusion therapy in AMI patients. Conclusion Phospho-Ser 9 -GSK-3β plays a critical role in intracellular signal mediated interventions that protect cardiomyocytes from ischemia/ reperfusion-induced necrosis. The mechanism of GSK-3β phosphorylation differs depending on the receptor activated by the intervention, but inactivation of GSK-3β by phosphorylation at Ser 9 elevates the threshold for mptp opening, which reduces myocyte necrosis. Although the mechanism by which phospho-ser 9 -GSK-3β elevates the threshold for mptp opening is unclear, suppression of ANT CyP-D interaction by binding of phospho-ser 9 -GSK-3β to ANT and reduction in GSK-3β mediated phosphorylation of p53 may be involved. The level of phospho-ser 9 -GSK-3β at the time of reperfusion is a determinant of infarct size, and GSK-3β inhibitors are promising as agents for myocardial salvage in patients with AMI. References 1 Jope RS, Yuskaitis CJ, Beurel E. Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics. Neurochem Res. 2007;32:577 595. 2 Cohen P, Goedert M. GSK3 inhibitors: development and therapeutic potential. Nat Rev Drug Discov. 2004;3:479 487. 3 Mora A, Sakamoto K, McManus EJ, Alessi DR. Role of the PDK1-PKB-GSK3 pathway in regulating glycogen synthase and glucose uptake in the heart. FEBS Lett. 2005;579:3632 3638. 4 Sugden PH, Fuller SJ, Weiss SC, Clerk A. Glycogen synthase kinase 3 (GSK3) in the heart: a point of integration in hypertrophic signalling and a therapeutic target? A critical analysis. Br J Pharmacol. 2008;153 Suppl 1:S137 S153. 5 Juhaszova M, Zorov DB, Kim SH, Pepe S, Fu Q, Fishbein KW, et al. Glycogen synthase kinase-3β mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 2004;113:1535 1549. 6 Tong H, Imahashi K, Steenbergen C, Murphy E. Phosphorylation of glycogen synthase kinase-3beta during preconditioning through a phosphatidylinositol-3-kinase dependent pathway is cardioprotective. Circ Res. 2002;90:377 379. 7 Gross ER, Hsu AK, Gross GJ. Opioid-induced cardioprotection occurs via glycogen synthase kinase-3β inhibition during reperfusion in intact rat hearts. Circ Res. 2004;94:960 966. 8 Gomez L, Paillard M, Thibault H, Derumeaux G, Ovize M. Inhibition of GSK3β by postconditioning is required to prevent opening of the mitochondrial permeability transition pore during reperfusion. Circulation. 2008;117:2761 2768. 9 Nishihara M, Miura T, Miki T, Sakamoto J, Tanno M, Kobayashi H, et al. Erythropoietin affords additional cardioprotection to preconditioned hearts by enhanced phosphorylation of glycogen synthase kinase-3β. Am J Physiol Heart Circ Physiol. 2006;291: H748 H755. 10 Nishihara M, Miura T, Miki T, Tanno M, Yano T, Naitoh K, et al. Modulation of the mitochondrial permeability transition pore complex in GSK-3β-mediated myocardial protection. J Mol Cell Cardiol. 2007;43:564 570.

166 T Miura et al 11 Piper HM, Abdallah Y, Schäfer C. The first minutes of reperfusion: a window of opportunity for cardioprotection. Cardiovasc Res. 2004;61:365 371. 12 Rodriguez-Sinovas A, Abadallah Y, Piper HM, Garcia-Dorado D. Reperfusion injury as a therapeutic challenge in patients with acute myocardial infarction. Heart Fail Rev. 2007;12:207 216. 13 Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N Engl J Med. 2007;357:1121 1135. 14 Matsumoto T, Miura T, Miki T, Genda S, Shimamoto K. Blockade of the Na + -Ca 2+ exchanger is more efficient than blockade of the Na + -H + exchanger for protection of the myocardium from lethal reperfusion injury. Cardiovasc Drugs Ther. 2002;16:295 301. 15 Förster K, Paul I, Solenkova N, Staudt A, Cohen MV, Downey JM, et al. NECA at reperfusion limits infarction and inhibits formation of the mitochondrial permeability transition pore by activating p70s6 kinase. Basic Res Cardiol. 2006;101:319 326. 16 Vinten-Johansen J, Zhao Z-Q, Jiang R, Zatta AJ, Dobson GP. Preconditioning and postconditioning: innate cardioprotection from ischemia-reperfusion injury. J Appl Physiol. 2007;103: 1441 1448. 17 Miura T. Myocardial Infarction. In: Sperelakis N, editor. Physiology and pathophysiology of the heart, 3rd ed. Norwell: Kluwer Academic Publisher; 1994. p. 1125 1145. 18 Komuro I, Ohtsuka M. Forefront of Na + /Ca 2+ exchange studies: Role of Na + /Ca 2+ exchanger lessons from knockout mice. J Pharmacol Sci. 2004;96:23 26. 19 Inserte J, Gacrcia-Dorado D, Hernando V, Barba I, Soler-Soler J. Ischemic preconditioning prevents calpain-mediated impairment of Na + /K + -ATPase activity during early reperfusion. Cardiovasc Res. 2006;70:364 373. 20 Halestrap AP, Clarke SJ, Javadov SA. Mitochondrial permeability transition pore opening during myocardial reperfusion-a target for cardioprotection. Cardiovasc Res. 2004;61:372 385. 21 Crompton M. Mitochondrial intermembrane junctional complexes and their role in cell death. J Physiol. 2000;529 Pt 1:11 21. 22 Griffiths EJ, Halestrap AP. Mitochondrial non-specific pores remain closed during cardiac ischaemia, but open upon reperfusion. Biochem J. 1995;307:93 98. 23 Petronilli V, Miotto G, Canton M, Brini M, Colonna R, Bernardi P, et al. Transient and long-lasting opening of the mitochondrial permeability transition pore can be monitored directly in intact cells by changes in mitochondrial calcein fluorescence. Biophys J. 1999;76:725 734. 24 Kim JS, Jin Y, Lemasters JJ. Reactive oxygen species, but not Ca 2+ overloading, trigger ph- and mitochondrial permeability transition-dependent death of adult rat cardiomyocytes after ischemia-reperfusion. Am J Physiol Heart Circ Physiol. 2006; 290:H2024 H2034. 25 Javadov SA, Clarke S, Das M, Griffiths EJ, Lim KH, Halestrap AP. Ischaemic preconditioning inhibits opening of mitochondrial permeability transition pores in the reperfused rat heart. J Physiol. 2003;549:513 524. 26 Argaud L, Gateau-Roesch O, Muntean D, Chalabreysse L, Loufouat J, Robert D, et al. Specific inhibition of the mitochondrial permeability transition prevents lethal reperfusion injury. J Mol Cell Cardiol. 2005;38:367 374. 27 Basso E, Fante L, Fowlkes J, Petronilli V, Forte MA, Bernard P. Properties of the permeability transition pore in mitochondria devoid of cyclophilin D. J Biol Chem. 2005;280:18558 18561. 28 Clarke SJ, McStay GP, Halestrap AP. Sanglifehrin A acts as a potent inhibitor of the mitochondrial permeability transition and reperfusion injury of the heart by binding to cyclophilin-d at a different site from cyclosporin A. J Biol Chem. 2002;277: 34793 34799. 29 Khaspekov L, Friberg H, Halestrap A, Viktorov I, Wieloch T. Cyclosporin A and its nonimmunosuppressive analogue N-Me- Val-4-cyclosporin A mitigate glucose/ oxygen deprivationinduced damage to rat cultured hippocampal neurons. Eur J Neurosci. 1999;9:3194 3198. 30 Li Y, Johnson N, Capano M, Edwards M, Crompton M. Cyclophilin-D promotes the mitochondrial permeability transition but has opposite effects on apoptosis and necrosis. Biochem J. 2004;383:101 109. 31 Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, Hambleton MA, et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature. 2005;434:658 662. 32 Nakagawa T, Shimizu S, Watanabe T, Yamaguchi O, Otsu K, Yamagata H, et al. Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature. 2005;434:652 658. 33 Lim SY, Davidson SM, Hausenloy DJ, Yellon DM. Preconditioning and postconditioning: the essential role of the mitochondrial permeability transition pore. Cardiovasc Res. 2007;75: 530 535. 34 Feng J, Lucchinetti E, Ahuja P, Pasch T, Perriard JC, Zaugg M. Isoflurane postconditioning prevents opening of the mitochondrial permeability transition pore through inhibition of glycogen synthase kinase 3beta. Anesthesiology. 2005;103:987 995. 35 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;61:448 460. 36 Watcharasit P, Bijur GN, Song L, Zhu J, Chen X, Jope RS. Glycogen synthase kinase-3β (GSK3β) binds to and promotes the actions of p53. J Biol Chem. 2003;278:48872 48879. 37 Venkatapuram S, Wang C, Krolikowski JG, Weihrauch D, Kersten JR, Warltier DC, et al. Inhibition of apoptotic protein p53 lowers the threshold of isoflurane-induced cardioprotection during early reperfusion in rabbits. Anesth Analg. 2006;103: 1400 1405. 38 Tsang A, Hausenloy DJ, Mocanu MM, Carr RD, Yellon DM. Preconditioning the diabetic heart: the importance of Akt phosphorylation. Diabetes. 2005;54:2360 2364. 39 Miki T, Miura T, Tsuchida A, Nakano A, Hasegawa T, Fukuma T, et al. Cardioprotective mechanism of ischemic preconditioning is impaired by postinfarct ventricular remodeling through angiotensin II type 1 receptor activation. Circulation. 2000;102: 458 463. 40 Miki T, Miura T, Tanno M, Sakamoto J, Kuno A, Genda S, et al. Interruption of signal transduction between G protein and PKC-ε underlies the impaired myocardial response to ischemic preconditioning in postinfarct remodeled hearts. Mol Cell Biochem. 2003;247:185 193. 41 Miki T, Miura T, Yano T, Takahashi A, Sakamoto J, Tanno M, et al. Alteration in erythropoietin-induced cardioprotective signaling by postinfarct ventricular remodeling. J Pharmacol Exp Ther. 2006;317:68 75.

GSK-3β in Cardioprotection 167 42 Miki T, Tanno M, Yano T, Satoh T, Hotta H, Ohori K, et al. Modification of Akt/ GSK-3β signaling and mitochondrial GSK-3β by ER stress underlies failure of erythropoietin to protect diabetic hearts. [Abstrac] Circulation. 2007;116:II-164. 43 Miura T, Yellon DM, Hearse DJ, Downey JM. Determinants of infarct size during permanent occlusion of a coronary artery in the closed chest dog. J Am Coll Cardiol. 1987;9:647 654. 44 Miura T, Yoshida S, Iimura O, Downey JM. Dobutamine modifies myocardial infarct size through supply-demand balance. Am J Physiol. 1988;254:H855 H861. 45 Miura T, Miki T. Limitation of myocardial infarct size in the clinical setting: current status and challenges in translating animal experiments into clinical therapy. Basic Res Cardiol. 2008;103:501 513. 46 Barillas R, Friehs I, Cao-Danh H, Martinez JF, del Nido PJ. Inhibition of glycogen synthase kinase-3β improves tolerance to ischemia in hypertrophied hearts. Ann Thorac Surg. 2007;84: 126 133.