用户名: 密码: 验证码:
糖尿病对心肌缺血后适应预防心肌再灌注损伤作用的影响及药物干预
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
心肌缺血后适应(myocardial ischemic postconditioning, MIPO)是在再灌注开始立刻给予短暂、多次、重复的心肌缺血与再灌注,然后再完全开放病变冠状动脉,能提高心肌对较长时间缺血的耐受性,减轻再灌注损伤以保护心肌。
     本研究将观察缺血后适应在正常大鼠及糖尿病大鼠中发挥预防心肌缺血再灌注损伤作用的异同,明确缺血后适应保护的作用机制,并探讨在病理状态下如何通过药物干预恢复或唤醒被弱化或消失的缺血后适应保护作用。
     研究结果表明:缺血后适应对于正常大鼠心肌再灌注损伤有保护作用,表现为心肌梗死面积减小及心肌酶释放减少、心脏血流动力学指标明显改善。PI3K-Akt信号通路在保护作用机制中起重要作用,免疫组化和western印迹分析显示缺血后适应的磷酸化Akt和GSK-3β表达增强。缺血后适应对于糖尿病大鼠离体心脏无保护作用,其磷酸化Akt、GSK-3表达减弱。糖尿病大鼠补充外源性锌(Zinc)后,缺血后适应预防再灌注损伤的作用得以恢复或唤醒,血流动力学指标改善和Akt、GSK-3β磷酸化水平增强。表明锌(Zinc)作为一种GSK-3β的直接或间接的抑制剂,可通过增强Akt磷酸化水平或直接抑制GSK-3β活性,使再灌注损伤救援激酶信号通路级联反应中下游物质GSK-3β磷酸化水平增高而失活,进而抑制心肌细胞线粒体通透性转换孔(mPTP)过度开放,保护缺血心肌免于再灌注损伤。
Coronary heart disease (CHD) is the leading cause of death in the world and represents one of the major burdens on healthcare systems today. Targeting those strategies that limit the damage sustained as a result of a lethal ischemic insult has been a major goal for many years.Reperfusion is the definitive treatment to salvage ischemic myocardium from infarction. A primary determinant of infarct size is the duration of ischemia. In myocardium that has not been irreversibly injured by ischemia, reperfusion induces additional injury in the area at risk. The heart has potent innate cardioprotective mechanisms against ischemia-reperfusion that reduce infarct size and other presentations of postischemic injury. In 1986, Murry et al. first introduced the concept of ischemic preconditioning (IPR) in which repetitive brief periods of ischemia protected the myocardium from a subsequent longer ischemic insult. Preconditioning succeeded in significantly reducing infarct size, preserve vascular endothelial function, and reduce apoptosis. Although preconditioning has been clinically successful, its use as a clinical cardioprotective strategy to attenuate the pathophysiological consequences (i.e., arrhythmias and infarction) of ischemia-reperfusion injury is limited by the inability to predict the onset of ischemia.
     In 2003, Zhao et al first described that brief episodes of ischemia performed at the onset of reperfusion after a prolonged ischemia provided a powerful anti necrotic protection, named“Postconditioning”(IPO), IPO defined as brief intermittent cycles of ischemia alternating with reperfusion applied after the ischemic event, has been shown to reduce infarct size, in some cases equivalent to that observed with IPO. Although there are similarities in mechanisms of cardioprotection by these two interventions, there are key differences that go beyond simply exerting these mechanisms before or after ischemia. On the other hand, IPO is applied at the point of service in the hospital, where and when reperfusion is initiated. Initial clinical studies are in agreement with the success and extent to which IPO reduces infarct size and myocardial injury, even in the presence of multiple comorbidities.
     Postconditioning has been shown to inhibit the mitochondrial permeability transition pore. Hence, postconditioning marshals a variety of endogenous mechanisms that operate at numerous levels and target a broad range of pathological mechanisms. The mechanism IPO protect of ischemic myocardial reperfusion injury is, After reflow, opening of the mitochondrial permeability transition pore (mPTP) has been involved in lethal reperfusion injury. Recent study has demonstrated that serine9 phosphorylation of GSK-3βis required for cardioprotection by postconditioning and likely acts by inhibiting opening of the mPTP at the time of reperfusion in a CypD-independent way .Postconditioning inhibits opening of the mPTP and provides a powerful antiischemic protection.Clinical studies in patients with acute myocardial infarction have demonstrated that postconditioning was effective in reducing infarct size. Postconditioning indirectly supports the concept of reperfusion injury in animal models of ischaemia–reperfusion and in patients, and exerts cardioprotection.We studied cardioprotection of IPO indiabetic heart existence or no, and how to wake-up and recovery cardioprotection of IPO in diabetic heart.
     Given the prevalence of ischaemic heart disease in diabetic patients, whether the diabetic heart is amenable to ischaemic postconditioning is an important unanswered question. Previous studies have demonstrated that postconditioning signals through the PI3K-Akt kinase pathway. Other studies have documented impaired PI3K-Akt signalling in the diabetic myocardium. We hypothesised that due to impaired signaling through the PI3K-Akt pathway, postconditioning does not protect the diabetic heart. Hearts isolated from the in-bred lean model of type II diabetes (High-fat diet+STZ induced rat) were randomised to either control or ischaemic postconditioning (IPost). Control hearts were mounted on a Langendorff apparatus and subjected to 30 min of myocardial ischaemia followed by either: (a) 10 min of reperfusion, at the end of which the myocardium at risk was analysed for Akt phosphorylation or (b) 120 min of reperfusion at the end of which myocardial infarct size was determined using tetrazolium staining. Hearts treated by IPost were subjected to 10 s episodes of alternate myocardial ischaemia/reperfusion applied at the end of the 30 min ischaemic period. Treatment with IPost did not reduce myocardial infarct size (45.65±4.8% in control versus 50.12±3.7% in IPost diabetic hearts; N = 10/group; P = NS). There was no difference in Akt phosphorylation between the two groups (51.5±4.6arbitrary units in control versus 54.1±4.2 arbitrary units in IPost diabetic hearts; N = 10/group; P = NS). And no difference in GSK-3βphosphorylation between the two groups (47.2±4.1arbitrary units in control versus 51.4±3.7 arbitrary units in IPost diabetic hearts; N = 10/group; P = NS). And, no improvement in hemodynamic parameters, include the HR, LVSP and +dp/dtmax. Exogenous zinc gived intraperitoneal injection in diabetic rats (ZnSO4 solution,50mg/Kg),and the action of IPO can be wake-up and recovery.show that improvement in hemodynamic parameters,and infart size reduced compared with the IPost heart of diabetic rats (31.10±3.4% VS 50.12±3.7%, p<0.01, n=10), There was Significant difference in Akt and GSK-3βphosphorylation between the two groups (67.6±3.4arbitrary units in Zinc+DM versus 54.1±4.2 arbitrary units in IPost diabetic hearts; N = 10/group; P <0.01 in Akt and 83.8±3.7arbitrary units in Zinc+DM versus 51.4±3.2 arbitrary units in IPost diabetic hearts; N = 10/group; P<0.01 in GSK-3β).
     In conclusion, we have demonstrated that postconditioning was effective in reducing infarct size. Postconditioningpostconditioning does not protect the diabetic heart which may be due to insufficient Akt and GSK-3βactivation. Exogenous zinc gived in diabetic rats the action of IPO can be wake-up and recovery.This finding may have important implications on the utilisation of this cardioprotective intervention in diabetic patients with ischaemic heart disease. IPO increases phosphorylation of GSK-3βthrough a PI3-kinase–PKB-dependent pathway, since wortmannin blocks the IPO-induced phosphorylation of GSK-3β. The GSK-3βinhibitors, Zinc, mimices the cardioprotective effect of IPO, suggesting that inhibition of GSK-3βis protective in IPO. We conclude that GSK-3βis involved in the cardioprotective effect of IPO through a PI3-kinase–dependent pathway. These data indicate that pharmacological modulation of GSK-3βactivity could have utility for protecting the heart against ischemic injury.
引文
[1] Zhao ZQ, Corvera JS, Halkos ME, Kerendi F, Wang NP, Guyton RA, Vinten-Johansen J. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. Am J Physiol Heart Circ Physiol. 2003;285:H579–H588.
    [2] Kin H, Zhao ZQ, Sun HY, Wang NP, Corvera JS, Halkos ME, Kerendi F, Guyton RA, Vinten-Johansen J. Postconditioning attenuates myocardial ischemia-reperfusion injury by inhibiting events in the early minutes of reperfusion. Cardiovasc Res. 2004;62:74–85.
    [3] Heusch G. Postconditioning: old wine in a new bottle? J Am Coll Cardiol 2004;44:1111-1112.
    [4] Matsumura K, Jeremy RW, Schaper J, et al. Progression of myocardial necrosis during reperfusion of ischemic myocardium Circulation. 1998, 97: 7952804.
    [5] James L. Park and Benedict R. Lucchesi Mechanisms of myocardial reperfusion injury Ann Thorac Surg 1999;68:1905-1912
    [6] Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium.Circulation, 1986,74(5):1124-1136.
    [7] Thourani VH, Nakamura M, Duarte IG, Bufkin BL, ZhaoZQ, Jordan JE, Shearer ST, Guyton RA, and Vinten-Johansen J. Ischemic preconditioning attenuates postischemic coronary artery endothelial dysfunction in a model of minimally invasive direct coronary artery bypass. J Thorac Cardiovasc Surg 117: 383–389, 1999.
    [8] Tomai F, Crea F, Chiariello L, and Gioffre PA. Ischemic preconditioning in humans: models, mediators, and clinical relevance. Circulation 100: 559–563, 2001.
    [9] Toyoda Y, Di Gregorio V, Parker RA, Levitsky S, andMcCully JD. Anti-stunning and anti-infarct effects of adenosine-enhanced ischemic precondintioning. Circulation 102:III-326–III-331, 2000.
    [10] Lim SY, Davidson SM, Hausenloy DJ, Yellon DM Preconditioning and postconditioning: the essential role of the mitochondrial permeability transition pore Cardiovasc Res. 2007 Aug 1;75(3):530-535.
    [11] Yellon DM, OpieLH. Postconditioning for protection of the infracting heart Lancet, 2006,367(9509):456-458.
    [12] Dow J, Bhandari A, Kloner RA. The mechanism by which ischemic postconditioning reduces reperfusion arrhythmias in rats remains elusive J Cardiovasc Pharmacol Ther. 2009 Jun;14(2):99-103.
    [13] Vinten-Johansen J, Zhao ZQ, Zatta AJ, Kin H, Halkos ME, Kerendi F. Postconditioning--A new link in nature's armor against myocardial ischemia-reperfusion injury Basic Res Cardiol. 2005 Jul;100(4):295-310.
    [14] Zweier JL. Measurement of superoxide-derived free radicals in the reperfused heart: evidence for a free radical mechanism of reperfusion injury. J Biol Chem 1988;263:1353-7.
    [15] Hearse DJ, Humphrey SM, Chain EB.Abrupt reoxygenation of the anoxic potassium-arrested perfused rat heart: a study of myocardial enzyme release. J Mol Cell Cardiol 1973;5:395-407.
    [16] Zweier JL, Talukder MA. The role of oxidants and free radicals in reperfusion injury. Cardiovasc Res 2006;70:181-90.
    [17] Piper HM, Garcia-Dorado D, Ovize M. A fresh look at reperfusion injury. Cardiovasc Res 1998;38:291-300.
    [18] Vinten-Johansen J. Involvement of neutrophils in the pathogenesis of lethal myocardial reperfusion injury. Cardiovasc Res 2004;61:481-97.
    [19] 19. Christopher P. Baines The mitochondrial permeability transition pore and ischemia-reperfusion injury Basic Res Cardiol (2009) 104:181–188
    [20] Hausenloy DJ, Ong SB, Yellon DM. The mitochondrial permeability transition pore as a target for preconditioning and postconditioning Basic Res Cardiol (2009) 104:189–202
    [21] Honda HM, Ping P. Mitochondrial permeability transition in cardiac cell injury and death. Cardiovasc Drugs Ther. 2006 Dec;20(6):425-32.
    [22] Lemasters JJ, Theruvath TP, Zhong Z, Nieminen AL. Mitochondrial calcium and the permeability transition in cell death. Biochim Biophys Acta. 2009 Nov;1787(11):1395-401
    [23] Piper HM, Abdallah Y, Sch鋐er C. The first minutes of reperfusion:A window of opportunity for cardioprotection. Cardiovasc Res 2004; 61: 365– 371.
    [24] 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 Res2006; 70: 364– 373.
    [25] Varadarajan SG, An J, Novalija E, Smart SC, Stowe DF. Changes in[Na(+)](i), compartmental [Ca(2+)], and NADH with dysfunctionafter global ischemia in intact hearts. Am J Physiol Heart Circ Physiol 2001; 280:H280–H293.
    [26] Tanno M, Miura T. Protecting ischemic hearts by modulation of SR calcium handling. Cardiovasc Res 2007;75:453–454.
    [27] Inserte J, Garcia-Dorado D, Ruiz-Meana M, Padilla F, Barrabés JA,Pina P, et al. Effect of inhibition of Na(+)/ Ca(2+) exchanger at the time of myocardial reperfusion on hypercontracture and cell death. Cardiovasc Res 2002;55:739–748.
    [28] Halestrap AP, Clarke SJ, Javadov SA. Mitochondrial permeability transition pore opening during myocardial reperfusion-a target for cardioprotection. Cardiovasc Res 2004; 61: 372– 85.
    [29] 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.
    [30] Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, HambletonMA, et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 2005;434:658–662.
    [31] Simpson PJ, Todd RF III, Fantone JC, Mickelson JK, Griffin JD, Lucchesi BR. Reduction of experimental myocardial reperfusion injury by a monoclonal antibody (anti-Mo1, anti-CD11b) that inhibits leukocyte adhesion. J Clin Invest 1988;81:624–9.
    [32] Ma XL, Tsao PS, Lefer AM. Antibody to CD-18 exerts endothelial and cardiac protective effects in myocardial ischemia and reperfusion. J Clin Invest 1991;88:1237-43.
    [33] 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.
    [34] Halestrap AP. What is the mitochondrial permeability transition pore? J Mol CellCardiol. 2009 Jun;46(6):821-31.
    [35] 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. Sep 18 2008.
    [36] Tsujimoto Y, Nakagawa T, Shimizu S. Mitochondrial membrane permeability transition and cell death. Biochim Biophys Acta. 2006 Sep-Oct; 1757(9-10):1297-300.
    [37] Beutner G. et al. Complexes between porin, hexokinase,mitochondrial creatine kinase and adenylate translocator display properties of the permeability transition pore. Implication for regulation of permeability transition by the kinases. Biochim. Biophys. Acta. 1998.1368: 7–18.
    [38] Crompton M. The mitochondrial permeability transition pore and its role in cell death. Biochem. J. 1999.341:233–249.
    [39] Kroemer, G, L. Galluzzi & C. Brenner. Mitochondrial membrane permeabilization in cell death. Physiol.Rev. 2007.87: 99–163.
    [40] Juhaszova M, Wang S, Zorov DB, Nuss HB, Gleichmann M, Mattson MP, et al. The identity and regulation of the mitochondrial permea bility transition pore: Where the known meets the unknown. Ann N Y Acad Sci 2008; 1123:197–212.
    [41] Derek M. Yellon, D.Sc., and Derek J. Hausenloy, Myocardial Reperfusion Injury N Engl J Med 2007;357:1121-35.
    [42] Halestrap A P, McStay G P, Clarke S J.The permeability transition pore complex:another view Biochimie, 2002, 84 (2-3):153-166.
    [43] Scorrano L, Ashiya M, Buttle K, et al. A distinct pathway remodels mitochondrial cristae and mobilizes cytochrome c during apoptosis .Dev Cell, 2002,2 (1):55-67.
    [44] Argaud L, Gateau-Roesch O, Chalabreysse L, Gomez L, Loufouat J,Thivolet-Bejui F, Robert D, Ovize M. Preconditioning delays Ca2+-induced mitochondrial permeability transition. Cardiovasc Res. 2004;61:115–122.
    [45] Argaud L,Gateau-Roesch O,Raisky O,Loufouat J,RobertD, OvizeM. Post- conditioning inhibits mitochondrial permeability transition. Circulation 2005;111:194-197
    [46] Speechly-Dick ME, Grover GJ, Yellon DM Does ischemic preconditioning in thehuman involve protein kinase C and the ATP-dependent K+ channel? Studies of contractile faction after simulated ischemic in an atrail in vitro model. Circ Res, 1995,77:1030~1035
    [47] Sommerschild HT, Kirkeboen KA. Preconditioning - endogenous defence mechanisms of the heart Acta Anaesthesiol Scand, 2002, 46(2):123-137..
    [48] Hugh B. Martin, MD, and Chris L. Walter, Preconditioning: An Endogenous Defense Against the Insult of Myocardial lschemia Anesth Analg 1996;83:639-645
    [49] Tsai BM, Wang M, March KL, et al. Preconditioning: evolution of basic mechanisms to potential therapeutic strategies .Shock, 2004, 21(3):195-209
    [50] Garlid KD, Paucek P, Yarov-Yarovoy V, et al. Cardioprotective effect of diazoxide and its interaction with mitochondrial ATPsensitive K+ channels. Possible mechanism of cardioprotection [J]. Circ Res, 1997, 81(6):1072-1082.
    [51] Pain T, Yang XM, Critz SD, et al. Opening of mitochondrial K (ATP) channels triggers the preconditioned state by generating free radicals. Circ Res, 2000, 87(6):460-466.
    [52] Kokoszka JE, Waymire KG, Levy SE, Sligh JE, Cai J, Jones DP, etal. The ADP/ATP translocator is not essential for the mitochondrial permeability transition pore. Nature 2004; 427: 461– 465.
    [53] Yellon DM, Baxter GF.A "second window of protection" or delayed preconditioning phenomenon: future horizons for myocardial protection? J Mol Cell Cardiol, 1995, 27(4):1023-1034.
    [54] Okamoto F, Allen BS, Buckberg GD, Bugyi H, Leaf J. Reperfusion conditions: importance of ensuring gentle versus sudden reperfusion during relief of coronary occlusion. J Thorac Cardiovasc Surg. 1986;92: 613–620.
    [55] Vinten-Johansen J, Lefer DJ, Nakanishi K, Johnston WE, Brian CA, Cordell RA. Controlled coronary hydrodynamics at the time of reperfusion reduces postischemic injury. Coron Artery Dis. 1992;3:1081–1093.
    [56] Sato H, Jordan JE, Zhao ZQ, Sarvotham SS, Vinten-Johansen J. Gradual reperfusion reduces infarct size and endothelial injury but augments neutrophil accumulation. Ann Thorac Surg. 1997;64:1099–1107.
    [57] Zweier JL, Flaherty JT, Weisfeldt ML. Direct measurement of free radicalsgenerated following reperfusion of ischemic myocardium. Proc Natl Acad Sci U S A. 1987;84:1404–1407.
    [58] Tsao PS, Aoki N, Lefer DJ, Johnson G III, Lefer AM. Time course of endothelial dysfunction and myocardial injury during myocardial ischemia and reperfusion in the cat. Circulation. 1990;82:1402–1412.
    [59] Piper HM, Schafer AC. The first minutes of reperfusion: a window of opportunity for cardioprotection. Cardiovasc Res. 2004;61:365–371.
    [60] 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
    [61] Hausenloy DJ, Tsang A, Yellon DM The Reperfusion Injury Salvage Kinase Pathway: A Common Target for Both Ischemic Preconditioning and Postconditioning Trends Cardiovasc Med 2005;15:69–75
    [62] Vinten-Johansen J. Postconditioning: a mechanical maneuver that triggers biological and molecular cardioprotective responses to reperfusion. Heart Fail Rev 2007; 12: 235–244.
    [63] Zhu M, Feng J, Lucchinetti E, et al. Ischaemic postconditioning protects remodeled myocardium via the PI3K-PKB/Akt reperfusion injury salvage kinase pathway. Cardiovasc Res 2006; 72: 152–162.
    [64] Sivaraman V, Mudalgiri NR, Di Salvo C, et al. Postconditioning protects human atrial muscle through the activation of the RISK pathway. Basic Res Cardiol 2007; 102: 453–459.
    [65] Tsang A, Hausenloy DJ, Mocanu MM, et al. Postconditioning: a form of“modified reperfusion”protects the myocardium by activating the phosphatidylinositol 3-kinase-Akt pathway. Circ Res 2004; 95:230–232.
    [66] Yang XM, Proctor JB, Cui L, et al. Multiple, brief coronary occlusions during early reperfusion protect rabbit hearts by targeting cell signalling pathways. J Am Coll Cardiol 2004;44:1103–1110.
    [67] Hausenloy DJ, Yellon DM. Survival kinases in ischaemic preconditioning and postconditioning. Cardiovasc Res 2006; 70: 240–253.
    [68] Juhaszova M, Zorov DB, Kim SH, Pepe S, Fu Q, Fishbein KW, ZimanBD, Wang S, Ytrehus K, Antos CL, Olson EN, Sollott SJ. Glycogen synthase kinase-3beta mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 2004;113:1535–1549.
    [69] Bopassa JC, Ferrera R, Gateau-Roesch O, Couture-Lepetit E, Ovize M. PI3-kinase regulates the mitochondrial transition pore in controlled reperfusion and postconditioning. Cardiovasc Res. 2006;69:178–185.
    [70] Argaud L, Gateau-Roesch O, Raisky O, Loufouat J, Robert D, Ovize M. Postconditioning inhibits mitochondrial permeability transition. Circulation. 2005;111:194–197.
    [71] Gomez L, Paillard M, Thibault H, et al. Inhibition of GSK3beta by postconditioning is required to prevent opening of the mitochondrial permeability transition pore during reperfusion. Circulation 2008; 117: 2761–2768.
    [72] Staat P, Rioufol G, Piot C, Cottin Y, Tri Cung T, L’Huillier I, Aupetit JF, Bonnefoy E, Finet G, Andre-Fouet X, Ovize M. Postconditioning the human heart. Circulation. 2005;112:2143–2148.
    [73] Baxter GF, Burley DS. Reperfusion and calculated RISKs: pharmacological postconditioning of human myocardium Br J Pharmacol. 2008 Jan;153(1):1-3.
    [74] Yamashita K, Kajstura J, Discher DJ et al Reperfusion activated Akt kinase prevents apoptosis in transgenic mouse hearts overexpressing insulin-like growth factor-1. Circ Res 2001;88:609–614.
    [75] Hanlon PR, Fu P, Wright GL, Steenbergen C, Arcasoy MO, Murphy E. Mechanisms of erythropoietin-mediated cardioprotection during ischemia-reperfusion injury: role of protein kinase C and phosphatidylinositol 3-kinase signaling. FASEB J 200519:1323–1325.
    [76] Bullard AJ, Govewalla P, Yellon DM Erythropoietin protects the myocardium against reperfusion injury in vitro and in vivo. Basic Res Cardiol 2005;100:397–493.
    [77] Sanada S, Asanuma H, Minamino T et al Optimal windows of statin use for immediate infarct limitation: 5’-nucleotidase as another downstream molecule of phosphatidylinositol 3-kinase. Circulation 2004;110:2143–2149.
    [78] Wynne AM, Mocanu MM, Yellon DM Pioglitazone mimics preconditioning in the isolated perfused rat heart: a role for the prosurvival kinases PI3K and P42/44MAPK. J Cardiovasc Pharmacol 2005,46:817–822.
    [79] Bell RM, Clark JE, Hearse DJ, Shattock MJ Reperfusion kinase phosphorylation is essential but not sufficient in the mediation of pharmacological preconditioning: characterization in the bi-phasic profile of early and late protection. Cardiovasc Res 2007;73:153–163.
    [80] Haffner SM, Lehto S, Ronnemaa T, et al. Motality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med, 1998,339:229-234.
    [81] Juutilainen A, Lehto S, et al. Type 2 diabetes as a“coronary heart disease equivalent”: an 18 year prospective population based study in Finnish subjects. Diabetes Care2005,28:2901
    [82] Norhammar A, Malmberg K, Diderholm E.Diabetes mellitus: 50% hemolytic unit of complement major risk factor in unstable coronary artery disease even after consideration of the extent of coronary artery disease and benefits of revascularization.J Am Coll Cardiol,2004,43:585-591.
    [83] Ali Raza J, Movahed ACurrent concepts of cardiovascular diseases in diabetes mellitus. Int J Cardiol. 2003 Jun; 89(2-3):123-34.
    [84]国家"九五"攻关计划糖尿病研究协作组,中国12个地区中老年人糖尿病患病率调查,中华内分泌代谢杂志. 2002:18(4).280-285
    [85] Yu C, Chen Y, Cline GW, et al. Mechanism by whick fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol3-kinase activity in muscle. J Biol Chem 2002;277(52):50230-50236.
    [86] Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature. 1999;399:601– 605.
    [87] Zeng G, Nystrom FH, Ravichandran LV, Cong LN, Kirby M, Mostowski H, Quon MJ. Roles for insulin receptor, PI3-kinase, and Akt in insulinsignaling pathways related toproduction of nitric oxide in human vascular endothelial cells. Circulation. 2000;101:1539–1545.
    [88] Eckel RH, Wassef M, Chait A, Sobel B, Barrett E, King G, Lopes-Virella M, Reusch J,Ruderman N, Steiner G, Vlassara H. Prevention Conference VI: Diabetes and Cardiovascular Disease: Writing Group II: pathogenesis of atherosclerosis in diabetes. Circulation. 2002;105:e138–e143.
    [89] Matsumoto T, Wakabayashi K, Kobayashi T, Kamata K. Alterations in vascular endothelial function in the aorta and mesenteric artery in type II diabetic rats. Can J Physiol Pharmacol. 2004;82:175–182.
    [90] Katso R, Okkenhaug K, Ahmadi K, White S, Timms J, Waterfield MD. Cellular function of phosphoinositide 3-kinases: implications for development, homeostasis, and cancer. Annu Rev Cell Dev Biol. 2001;17:615–675
    [91] Derek J. Hausenloy, Andy Tsang, et al. Postconditioning does not protect the diabetic heart. ABSTRACTS / Journal of Molecular and Cellular Cardiology 2006;40: 920–1015.
    [92] Jope RS, Yuskaitis CJ, Beurel E. Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics. Neurochem Res. 2007;32:577–595
    [93] 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.
    [94] 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
    [95] 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.
    [96] 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.
    [97] 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.
    [98] Miki T, Miura T, Tanno M, Sakamoto J, Kuno A, Genda S, et al. Interruption of signal transduction between G protein and PKC-εunderlies the impaired myocardial response to ischemic preconditioning in postinfarct remodeled hearts.Mol Cell Biochem. 2003;247:185–193.
    [99] Miki T, Tanno M, Yano T, Satoh T, Hotta H, Ohori K, et al. Modification of Akt/GSK-3βsignaling and mitochondrial GSK-3βby ER stress underlies failure of erythropoietin to protect diabetic hearts.[Abstrac]Circulation. 2007;116:II-164.
    [100] Gross ER, Hsu AK, Gross GJ.Opioid-induced cardioprotection occurs via glycogen synthase kinase-3βinhibition during reperfusion in intact rat hearts.Circ Res. 2004;94:960–966.
    [101] 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.
    [102] Barillas R, Friehs I, Cao-Danh H, Martinez JF, del Nido PJ.Inhibition of glycogen synthase kinase-3βimproves tolerance to ischemia in hypertrophied hearts.Ann Thorac Surg. 2007;84:126–133.
    [103] 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:S137–S153.
    [104] Miura T, Nishihara M, and Miki T. Drug Development Targeting the Glycogen Synthase Kinase-3β(GSK-3β)-Mediated Signal Transduction Pathway: Role of GSK-3βin Myocardial Protection Against Ischemia/Reperfusion Injury J Pharmacol Sci 2009;109, 162-167.
    [105] Coghlan MP,Culbert AA,Cross DA. Selective small molecular inhibitors of glycogen synthase kinase-3 modulate glycogen metabolism and transcription.Chem Biol,2000,7:793-803
    [106] Marcus G,Friedrich H.Inhibitor of glycogen synthase kinase (GSK)3 promotes replication and survival of pancreatic beta cells J Biol Chem,2007,282:12030-12037
    [107] Klein PS, Melton DA. A molecular mechanism for the effect of lithium on development Proc Natl Acad Sci USA. 1996 August 6; 93(16): 8455–8459.
    [108] Krezel A, Hao Q, Maret W. The zinc/thiolate redox biochemistry of metallothionein and the control of zinc ion fluctuations in cell signaling. Arch Biochem Biophys 2007;463: 188–200.
    [109] Beyersmann D, Haase H. Functions of zinc in signaling, proliferation and differentiation of mammalian cells. Biometals 2001.14: 331–341.
    [110] An WL, Pei JJ, Nishimura T, Winblad B, Cowburn RF. Zinc-induced anti-apoptotic effects in SH-SY5Y neuroblastoma cells via the extracellular signal-regulated kinase 1/2. Brain Res Mol Brain Res 2005.135: 40–47.
    [111] Ilouz R, Kaidanovich O, Gurwitz D, Eldar-Finkelman H. Inhibition of glycogen synthase kinase-3_ by bivalent zinc ions: insight into the insulin-mimetic action of zinc. Biochem Biophys Res Commun 295: 102–106, 2002.
    [112] Jang Y, Wang H, Xi J, Mueller RA, Norfleet EA, Xu Z. NO mobilizes intracellular Zn2+ via cGMP/PKG signaling pathway and prevents mitochondrial oxidant damage in cardiomyocytes. Cardiovasc Res2007. 75: 426–433.
    [113] Miura T, Miki T, GSK-3β, a Therapeutic Target for Cardiomyocyte Protection Circ J 2009; 73:1184–1192.
    [114] Alonso M. Martinez A. GSK-3 Inhibitors: Discoveries and Developments. Current Medicinal Chemistry, Volume 11, Number 6, March 2004, pp. 755-763(9).
    [115] Chanoit G, Lee S, Xi J, Zhu M, McIntosh RA, Mueller RA, Norfleet EA, Xu Z. Exogenous zinc protects cardiac cells from reperfusion injury by targeting mitochondrial permeability transition pore through inactivation of glycogen synthase kinase-3beta. Am J Physiol Heart Circ Physiol. 2008 Sep;295(3):H1227-H1233.
    [116] Chimienti F, Aouffen M, Favier A, Seve M. Zinc homeostasis-regulating proteins: new drug targets for triggering cell fate. Curr Drug Targets 2003.4: 323–338.
    [117] Tosaki A, Engelman DT, Engelman RM, Das DK The evolution of diabetic response to ischemia/reperfusion and preconditioning in isolated working rat hearts. Cardiovasc Res 1996;31:526–536.
    [118] Kersten JR, Toller WG, Gross ER, Pagel PS, Warltier DC Diabetes abolishes ischemic preconditioning: role of glucose, insulin, and osmolality. Am J Physiol Heart Circ Physiol 2000;278:H1218–H1224.
    [119] del Valle HF, Lascano EC, Negroni JA Ischemic preconditioning protection against stunning in conscious diabetic sheep: role of glucose, insulin, sarcolemmal and mitochondrial KATP channels. Cardiovasc Res 2002;55:642–659.
    [120] Hassouna A, Loubani M, Matata BM, Fowler A, Standen NB, Galinanes M Mitochondrial dysfunction as the cause of the failure to precondition the diabetic human myocardium. Cardiovasc Res 2006;69:450–458.
    [121] Sekler I, Sensi SL, Hershfinkel M, Silverman WF. Mechanism and regulation of cellular zinc transport. Mol Med 2007.13:337–343.
    [122] Barthel A, Ostrakhovitch EA, Walter PL, Kampkotter A, Klotz LO. Stimulation of phosphoinositide 3-kinase/Akt signaling by copper and zinc ions: mechanisms and consequences. Arch Biochem Biophys 2007.463: 175–182.
    [123] Mocanu MM, Yellon DM. PTEN, the Achilles’heel of myocardial ischaemia/ reperfusion injury? Br J Pharmacol 2007.150: 833–838.
    [124] Wu W, Wang X, Zhang W, Reed W, Samet JM, Whang YE, Ghio AJ. Zinc-induced PTEN protein degradation through the proteasome pathway in human airway epithelial cells. J Biol Chem 278:28258–28263, 2003.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700