用户名: 密码: 验证码:
心肌缺血/再灌注损伤及钙调神经磷酸酶信号通路的调控作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
心肌缺血再灌注(I/R)损伤可产生心肌细胞凋亡和坏死,导致心肌梗死面积扩大、心室重塑和心力衰竭。本研究从心肌I/R损伤致细胞凋亡的钙超载机制入手,探讨了钙调神经磷酸酶(CaN)抑制剂FK506在心肌I/R损伤过程中的作用,揭示了CaN在心肌I/R损伤致心肌细胞凋亡的调控机制。以大鼠H9c2心肌细胞为研究对象,采用四甲基偶氮唑盐(MTT)法检测过氧化氢对心肌细胞的损伤作用,单细胞凝胶电泳法检测DNA损伤,吖啶橙/溴乙锭(AO/EB)双荧光染色和流式细胞术检测细胞凋亡;并建立大鼠H9c2心肌细胞凋亡的损伤模型,给予不同剂量FK506,采用流式细胞术检测细胞凋亡率,观察FK506对心肌细胞凋亡的影响。建立大鼠心肌I/R损伤模型为研究对象,给予FK506进行干预,观察I/R损伤及药物干预后心肌梗死面积,心律失常程度以及血压的变化,采用生物化学法检测心肌细胞脂质过氧化水平、组织酶CK和LDH活性变化,采用免疫组织化学法检测心肌细胞凋亡相关蛋白的表达,采用Western Blot方法检测CaN蛋白的表达情况。结果表明:H2O2可引起心肌细胞的DNA损伤,诱导细胞凋亡。FK506对H2O2诱导的大鼠H9c2心肌细胞凋亡具有抑制作用。FK506干预可缩小心肌梗死面积,减轻心肌I/R损伤。FK506可增加Bcl-2蛋白表达,抑制心肌细胞凋亡。因此,FK506通过线粒体途径诱导心肌I/R损伤所致的心肌细胞凋亡,CaN在I/R损伤中具有重要作用。抑制CaN活性对于抑制心肌I/R损伤,减少心肌细胞凋亡,改善心脏功能具有特别重要的意义。
After an acute ischemic heart disease, early and successful myocardial reperfusion is an effictive strategy for obtaining thoroughly coronary blood flow, saving the dying myocardiocyte and reducing the size of a myocardial infarct. Because of the uncertainty relates to the ischemic period and degree, myocardial ischemia/reperfusion injury caused by the restoration of coroary blood flow after an ischemic episode. This form of myocardial injury can induce cardiomyocyte apoptosis and necrosis, increase infarct size, remodel cardiac ventricle, then the ultmate incidence is cardiac failure. To observe H_2O_2-induced myocardial H9c2 cell apoptosis and pretective effect of inhibitor of calcineurin (CaN), a cell apoptotic model from rat myocardiocytes cell line H9c2 induced by hydrogen dioxide (H_2O_2) and a rat model of ischemia/reperfusion are established. These provide theoretical basis for study on CaN induced myocardiocyte apoptosis caused by ischemia/reperfusion indury and related mechanism.
     1.Toxicity of H_2O_2 on H9c2 cells
     1.1 The survival rate of H9c2 cell detected by MTT MTT assay was performed to detect the toxicity after 1, 3, 6, 12 and 24 h exposure to H_2O_2 (0, 50, 100, 200 and 400μmol/L). It was showed the survival rate decreased with the increase of dosage and duration of exposure to H_2O_2. After 6 h exposure to 200μmol/L H_2O_2,half of the H9c2 cells died. The change of survival rate was over 60%, with 24 h after exposure 400μmol/L H_2O_2. There was significant difference in survival rate between H_2O_2 group and control group(NC) (P < 0.05). The results suggest that cardiomyocytes are sensitive to H_2O_2-induced dosage.
     1.2 DNA damage of H9c2 cells
     Single cell gel electrophoresis (SCGE) assay was used to examine DNA damage of H9c2 cells 6 h after exposure to H_2O_2 (0, 50, 100, 200 and 400μmol/L). When exposed to 50, 100, 200 and 400μmol/L H_2O_2 for 6 h, there were significant difference from control group. The results suggest H_2O_2 could cause DNA damage of myocardiocytes in dose dependant way.
     1.3 Apoptosis detected by AO/EB double fluorescent staining assay There were distinctive changes on cell apoptosis observed under fluorescence microscope 1, 3, 6 and 12 h after exposure to H_2O_2 (0, 50, 100, 200 and 400μmol/L) by AO/EB double staining. The H9c2 cells occur obviously morphological changes in H_2O_2 -exposed groups, and the apoptotic rate was a significant increase compared with control group (P < 0.05), the highest group at 6 h after exposure to 100μmol/L H_2O_2. Whthin a prolong time exposure to it, the change of apoptotic rate is not obvious.
     1.4 Apoptosis detected by FCM with AnnexinⅤ/PI double fluorescent staining Cell apoptosis was observed by FCM 1, 6 and 24 h after exposure to H_2O_2 (0, 50, 100, 200 and 400μmol/L). The apoptotic rates were significantly increased in 100, 200 and 400μmol/L H_2O_2 at 6 and 24 h compared with control group(P < 0.05), the highest group at 24 h after exposure to 400μmol/L H_2O_2. The results suggest H_2O_2 could induce the H9c2 cell apoptosis in dose dependant and time dependant way.
     1.5 Membrane potential detected by FCM with Rhodamine 123 fluorescent prode
     Mitochondria membrane potential (Δψmt) was observed by FCM 1, 6 and 24 h after exposure to H_2O_2 (0, 50, 100, 200 and 400μmol/L).Δψmt showed the significant increase in H_2O_2 -exposed groups at 1 h compared with control group (P < 0.05). Whthin a prolong time exposure to it, the change ofΔψmt is observed. Compared with control group,Δψmt showed the significant increase in 50 and 100μmol/L H_2O_2 at 6 h, and the significant decrease in 200 and 400μmol/L H_2O_2 at 24h (P < 0.05). The results suggest H_2O_2 could damage mitochondria, decreaseΔψmt and induce the apoptosis in myocardial H9c2 cell.
     2. Effect of inhibitor of calcineurin on myocardial H9c2 cell apoptosis induced by H_2O_2
     The myocardial cell apoptosis models of rats were established, that is, myocardial H9c2 cells was exposed to 100μmol/L H_2O_2 at 6 h. FK506 were administerted to the model group at low dose (0.15μmol/L) and high dose (0.60μmol/L) levels, and the apoptotic and necrotic rates were detected by FCM with AnnexinⅤ/PI double fluorescent staining. Compared with 100μmol/L H_2O_2-exposed group at 1 h, the H_2O_2-induced apoptotic rate was significantly decreased in FK506 treated group (P < 0.05). The inhibitor of CaN shows protective effect on myocardial H9c2 cell apoptosis induced by H_2O_2, with dose-undependant manner.
     3. The protective effect on myocardial ischemia/reperfusion injury of inhibitor of CaN in rats
     3.1 A rat heart model of ischemia/reperfusion injury established Weigh and anesthetized the rat , then fix it on the operation table and removed the fur from the neck and the left chest. The trachea was intubated and arterial blood pressure was continuously monitored via a saline-filled catheter inserted into the common carotid artery. The chest was opened and the pericardium was incised. The heart was gently exteriorized by applying pressure on the right side of the chest, and a ligature wa placed around the left anterior descending coronary (LAD) occlusion 30 min and reperfusion 120 min. Then a rat model of myocardial ischemia/reperfusion injury was been established. Wistar rats were randomly divided into four groups: I/R group, FK506-I/R group (FK506 was intravenouosly injected 15 min before ischemia), Sham group (a ligature was placed around LAD and no occluded), FK506-Sham group (FK506 was intravenously injected , a ligature was placed around LAD and no occluded).
     3.2 Monitoring mean blood pressure, heart rate and electrocardiogram Mean blood pressure (MBP) was continuously monitored via an pressure transducer connected to a physiograph. Electrocardiogram and heart rate (HR) are measured by LeadⅡusing an electrocardiogram/rate coupler, both being analyzed by the BL-410 BioLab System. Four period of MBP and HR were selected (baseline, before occlusion, end-ischemia, end-reperfusion). Evaluation of arrhythmias were based on the description of the Lambeth Conventions. Total number of episodes for premature ventricular contraction (PVC), total duration for ventricular tachycardia (VT) and ventricular fibrillation (VF) were evaluated. The severity of arrythmias was quantified by a scoring system. During the ischemia period, MBP was a significant decrease in I/R group compared with in FK506-I/R group (P < 0.05). But during the reperfusion period, these two groups were no significant difference (P > 0.05). During the four period, the changes of HR were not obvious. Administration of FK506 before I/R significantly reduced the occurrence of PVC, VT, VF and duration of cardiac arrhythmia, and reduced the arrhythmia score (P > 0.05). The results suggest FK506 shows antiarrhythmic and cardioprotective effects in rat model of myocardial ischemia/reperfusion injury.
     3.3 Area at risk myocardial infarct size determination
     The area at risk (ischemic/reperfused region) and the infarct size were measured using Evans blue and 2,3,5-triphenyltetrazolium chloride (TTC) double staining method. The images of heart slices were captured by a camera and area of infarcted myocardium was digitally measured using Image-Pro plus. Area-at-risk (AAR)/left-ventricular area (LV)×100%, infarct zone (IS)/LV×100%, IS/AAR×100% were calculated. Compared with I/R group, IS/AAR in FK506-I/R group was singnificant decrease (P < 0.05). The result indicated FK506 signifivantly reduced myocardial ischemia/reperfusion injury.
     3.4 Assay of myocardial enzyme leakage
     At the end of reperfusion, blood samples were collected to measure the myocardial enzyme leakage, including creatinine kinase (CK) and lactate dehydrogenase (LDH). CK and LDH kits were applied to examine their activities. It was showed the activities of CK and LDH were siginificant decrease in FK506-I/R group compared with I/R group (P < 0.05).
     3.5 Lipid peroxidation of plasma examination
     SOD kit was applied to examine activities of SOD in rats plasma. It was showed the activities of SOD were significant decrease in both FK506-I/R and I/R groups, compared with Sham group (P < 0.05). There were no significant changes from FK506-I/R to I/R group(P > 0.05). TBA colorimetric method was applied to examine MDA contents. It was showed the MDA contents were significant increase in both FK506-I/R and I/R groups, compared with Sham group (P < 0.05). There were no significant changes from FK506-I/R to I/R group(P > 0.05).
     3.6 Changes of CaN enzyme activites
     CaN kit was applied to examine activites of CaN in myocardium tissues. The activities of CaN were significant decrease in FK506-I/R group compared with I/R group(P < 0.05).
     4. CaN induced myocardiocyte apoptosis caused by ischemia/reperfusion indury and related mechanism.
     4.1 Changes of expression of Bcl-2, Bax, and Caspase– 3 Immunocytochemical method was used to detect expression of Bax, Bcl-2 and Caspase-3. It could be observed under optical microscope that nucleus of positive expressed cells was dyed dark blue and cytoplasm or cellular membrane was dyed brown. Expression of Bcl-2 significantly increased in FK506-I/R group, and expression of Bax significantly increased in I/R group compared with Sham group(P < 0.05). The value of Bcl-2/Bax were a significant difference between FK506-I/R and I/R group(P < 0.05). Expression of Caspase-3 was a significant increase in FK506-I/R and I/R group compared Sham group (P < 0.05). The result suggests FK506 could depress myocardiocyte apoptosis.
     4.2 Changes of expression of CaN
     Western blot was applied to detect CaN activity, and the characteristic 62 kDa fragment of CaN was observed in four groups. The expression of CaN enhanced and got to the highest in I/R group, the lowest in FK506-Sham group.
     Generally, H_2O_2 could cause oxidative injury and DNA damage. It could also cause mitochondrial damage, then decrease mitochondrial mambrane potential. So H_2O_2 could induce the apoptosis on myocardial H9c2 cells. The inhibitor of CaN, FK506, could suppress H_2O_2-induced apoptosis on the H9c2 cells, and shows cardioprotective effect . FK506 could reduce the size of infarct zone on a rat model of myocardial ischemia/reperfusion injury, and increase the expression of Bcl-2 protein. So the inhibitor of CaN could suppress myocardiocyte apoptosis, the mechanism is related to CaN production and induced apoptosis by mitochondria signal transduction.
引文
[1] Jennings RB, Sommers HM, Smyth GA, et al. Myocardial necrosis induced by temporary occlusion of a coronary artery in the dog[J]. Arch pathol, 1960; 70: 68-78.
    [2] Gottlieb RA, Burleson KA, Kloneer RA, et al. Reperfusion injury induces apoptosis in rabbit cardiomyocytes[J]. Invest, 1994, 94 (4) :1621.
    [3] Gumina RJ,Buerger E,Eickmeier C, et al. Inhibition of the Na+/H+ exchanger confers greater cardioprotection against 90 minutes of myocardial ischemia than dichemic precongditioning in dogs[J]. Circulation 1999;100:2519-26.
    [4] Braunwald E, Kloner RA. The stunned myocardium: prolonged, postischemic ventricular dysfunction[J]. Circulation 1982; 66: 1146-9.
    [5] Ito H. No-reflow phenomenon and prognosis in patients with acute myocardial infarction[J]. Nat Clin Pract Cardiovasc Med 2006; 3: 499-506.
    [6] Manning AS, Hearse DJ. Reperfusion-induced arrhythmias: mechanisms and prevention[J]. Mol Cell Cardiol 1984; 16: 497-518.
    [7] Piper HM, Garcia-Dorado D, Ovize M. A fresh look at reperfusion injury[J]. Cardiovasc Res 1998; 38: 291-300.
    [8] Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium[J]. Circulation, 1986, 74: 1124.
    [9] Przyklenk K, Bauer B, Ovize M, et al. Regional ischemic preconditioning protects remote virgin myocardium from subsequent sustained coronary occlusion[J]. Circulation, 1993; 87: 893-99.
    [10] Olafsson B, Foman MB, Puett DW, et al. Reduction of reperfusion injury in the canine preparation by intracoronary adenosine: importance of the endothelium and the no-reflow phenomenon[J]. Circulation, 1987, 76: 1135-45.
    [11] McClanahan JB, Nao BS, Wolke LJ, et al. Brief renal occlusion and reperfusion reduces myocardial size in rabbit[J]. FASEB J,1993; 7: A118.
    [12] Zhao ZQ, Corvera JS, Halkos ME, et al. Inhibition of myocardial injury by ischemic postconditioning[J]. Am J Physiol Heart Circ Physiol, 2003; 285: H579-88.
    [13] Vinten- Johansen J. Reperfusion injury: idle curiosity or therapeutic vector? [J].Thromb Thrombolysis, 1997; 4: 59-61.
    [14] Staat P, Rioufol G, Piot C, et al. Postconditioning the human heart[J]. Circulation 2005; 112: 2143-8.
    [15] Mehta SR, Yusuf S, Diaz R, et al. Effect of glucose-insulin-potassium infusion on mortality in patients with acute ST-segment elevation myocardial infarction: the CREATE-ECLA randomized controlled trial[J]. JAMA 2005; 293: 437-46.
    [16] Canbaz S, Duran E, Ege T,et al. The effects of intracoronary administration of vitamin E on myocardial ischemia-reperfusion injury during coronary artery surgery[J]. Thorac Cardiovasc Surg, 2003, 51; 57-61.
    [17] Riley RD, Sato H, Zhao ZQ, et al. Recombination human complement C5a receptor antagonist reduces infarct size after surgical revascularization[J]. Thorac Cardiovasc Surg, 2000, 120: 350-58.
    [18]蒋立新.心脏保护研究与临床实践[J].实用医院临床杂志; 2004, 1 (2); 18-20.
    [19] Klein HH, Pich S, Lindert S, et al. Treatment of reperfusion injury with intracoronary calcium channnel antagonists and reuced coronary free calcium concentration in regionallu ischemic, reperfused porcine hearts[J]. J Am coll cardiol 1989; 13: 1395-401.
    [20] Zeymei U, Suryapranata H, Monassier JP,et al. The Na(+)/H(+)exchange inhibitor eniporide as an adjunct to early reperfusion therapy for acute myocardial infarction: results of the Evaluation of the Safety and Cardioprotective Effects of Eniporide in Acute Myocardial Infarction (ESCAMI) trial[J]. J Am Coll Cardiol 2001; 38: 1644-50.
    [21] Bǒr FW, Tzivoni D, Dirksen MT, et al. Results of the first clinical study of adjunctive CAldaret (MCC-135) in patients undergoing primary percutaneous coronary intervention for ST-Elevation Myocardial Infarction: the randomized multicentre CASTEMI study[J]. Eur Heart J 2006; 27: 2516-23.
    [22] Hausenloy DJ, Duchen MR, Yellon DM. Inhibiting mitochondrial permeability transition pore opening at reperfusion protects against ischemia-reperfusion injury[J]. Cardiovasc Res 2003; 60: 617-25.
    [23]陈凌.心肌缺血再灌注损伤与钙通道阻滞剂的保护作用[J].心血管病学进展. 1991, 12 (2): 96-100.
    [24]司良役.心肌再灌注损伤与钙超载、自由基及中性粒细胞[J].中国循环杂志, 1996, 11 (6): 382-3.
    [25] Hess ML, Manson NH. The role of oxygen free radical system in thecalcium paradox, the oxygen paradox and ischemia/reperfusion injury[J]. J Mol Cell Cardiol, 1984, 16: 969.
    [26] Dixon IM, Kaneko M, Hata T, et al. Alterations in cardial membrane Ca2+ transport during oxidative stress[J]. Mol Cell Biochem, 1990, 99: 125-33.
    [27] Otani H, Engelman RM, Rousou JA, et al. Cardiac performance during reperfusion improved by pretreatment with oxygen frdd-redical scavengers[J]. J Thorac Cardiovasc Surg, 1986, 91: 290.
    [28] Uchiyama T, Otani H, Okada T, et al. Nitric oxide induces caspase-dependent apoptosis and necorosis in neonatal rat cardiomyocytes[J]. J Mol Cell Cardiol, 2002, 34: 1049-61.
    [29] Gozal Y, Drenger B. Myocardial ischemic preconditioning decreases postischemic oxygen free radical production[J]. Anesthesiology, 2003, 98: 281-2.
    [30] Das S, Engelman RM, Maulik N, et al. Angiotensin preconditioning of the heart: evidence for redox signaling[J]. Cell Biochem Biophys, 2006, 44: 103-13.
    [31] Shen AC, Jennings RB. Kinetics of accumulation in acute myocardial ischemic injury[J]. Am J Pathol, 1972, 67: 441-7.
    [32] Huser J, Rechemacher CE, Blatter LA. Imaging the permeability pore transition in single mitochondria[J]. Biophys J 1998; 74(4): 2129-37.
    [33] Goldhaber JL, Qayyum MS. Oxygen free radicals and excitation-contraction coupling. Antioxid Redox Signal, 200, 2: 55-64.
    [34] Petrosilb G, Ruggiero FM, DiVenosa N, et al. Decreased complexⅢactivity in mitochondria isolated from rat heart subjected to ischemia and reperfusion: role of reactive oxygen species and cardiolipin[J]. FASEB J, 2003, 17: 714-6.
    [35] Das DK, Naogi A. Effets of superoxide amions the Na+-K+-ATP system in rat lung [J]. clin ph ysiol Biochem,1984, 2002, 2 (4): 328
    [36] Barry S, Oemar, Marcel R, et al. Reduced endotheial nitric oxide synthase exoression and producion in human atheroslerosis[J]. Circulation, 1998, 97: 2 494.
    [37]林函,王祥瑞.心肌缺血再灌注损伤中线粒体的作用[J].《国外医学》麻醉学与复苏分册, 2002, 23(5): 275-278.
    [38] Dreyer WJ, Smith CW, Michael LH, et al. Canine neutrophil activation by cardiac lymph obtained during reperfusion of ischemic myocardium[J]. Circ Res, 1989, 65: 1751-62
    [39] Royston D, Fleming JS, Desai JB, et al. Increased production ofperoxidation products associated with cardiac operations evidence for free radical generation[J]. J Thorac Cardiovasc Surg, 1986, 91: 759.
    [40] Georg M, Sherman MP, Bukbeng GD, et al. Role of L-arginine-nitrio oxido pathway in myocardial reoxygenation injury[J]. Am J Physiol, 1992, 262: 626.
    [41] Kawano H. Normal and abnormal conseqrences of apoptosis in the human heart: from postnatal morphogenesis to paroxymal arrthymias[J]. Jpn Heart J, 1994, 35(6): 745.
    [42] Scarabelli TM, Knight R, Atephanou A, et al. Clinical implications of apoptosis in sichemic myocardium[J]. Curr Probl Cardiol, 2006, 31(3):181-264.
    [43] Sdzner M, Rudiger HA, Adzner N. Transgenic mice overexperssing human are resistant to hepatic ischemia and reperfusion. J Hepatol, 2002, 36(2): 218-225.
    [44] Bursch W, Pafe S, Putz B, et al. Determination of the length of the histological stages of apoptosis hepatic foci of rats[J]. Cancigenesis, 1990, 11 (2): 847-851.
    [45] Wyllie AH, Kerr JFR, Currie AR. Cell death: the significance of apoptosis[J]. Int Rev Cytol, 1980, 68 (3): 251-253.
    [46] Oberhammer F, Wilson JW, Dive C, et al. Apoptotic death in epithelial cells: clevage of DNA to 300 and 50 kb fragments prior to in the absence of internuclaosomal fragmentation[J]. EMBO J, 1993, 12 (3): 3679-3682.
    [47] Gorczyca W, Gong J, Ardelt B, et al. The cell cycle related differences in susceptibility of HL260 cells to apoptosi sinduced by various antitumo agents[J]. Cancer Res, 1993, 53 (13): 3186-3192.
    [48] Ormerod MG, Sun XM, Brown D, et al. Quantification of apoptosis and necrosis by flow cytometry[J]. Acta Oncol, 1993, 32 (4): 417-424.
    [49] VanEngeland M, Nieland LJ, Ramaekers FC, et al. Annexin V-affinit assay:a review on an apoptosis detection system based on phosphatidyl serine exposure[J]. Cytometry, 1998, 31 (1): 129.
    [50] Span LFR, Pennings AHM, Vierwinden G, et al. The dynamic process if apoptosis analyzed by flow cytometry using Annexin-V/Propidium Iodide and a modified in situ end labeling technique[J]. Cytometry, 2002, 47: 24-31.
    [51] Negoesu A, Lorimier P, Labat MF, et al. In situ apoptotic cell labeling by the TUNEL method: improvement and evaluation on cell preparations[J]. J Histochem Cytochem Cytochem, 1996, 44 (3): 959-963.
    [52] Eskes R, Desagher S, Antonsson B, et al. Bid induces the oligomerization and insertion of Bax into the outer mitochondrial membrane[J]. Mol Cell Biol, 2000,20(3): 929-35.
    [53] Ashkenazi A, Dixit VM. Death receptors: signaling and modulation[J]. Science, 1998, 281(5381):1305-8.
    [54] Nakagawa T, zhu H, Morishima N, et al. Aoaf-1 oligomerizes into biologically active approximately 700-kDa and inactive approximately 1.4-kDa apoptosome complexes [J]. J Biol Chem, 2000, 275(9): 6067-70.
    [55] MoConkey DJ, Nutt LK. Calcium flux measurements in apoptosis J. Methods Cell Biol, 2001, 66:229-46.
    [56] Li H, Zhu H, Xu CJ, et al. Cleavage of BID by Caspase-8 mediates the mitochondrial damage in the Fas pathway of apoptosis [J]. Cell, 1998, 94(4): 491-501.
    [57] Grunenfelder J, Miniati DN, Murata S, et al. Upregulation of Bcl-2 through caspase-3 inhibition ameliorates ischemia/reperfusion unjury in rat cardiac allografts[J]. Circulation, 2001; 104: 1202-06.
    [58] Sujimoto Y, Cossman J, Jaffe E, et al. Involvement of the bcl-2 gene in human follicular lymphoma[J]. Science, 1985, 228 (4706): 1440-1443.
    [59]永锋,樊廷俊. Bcl-2家族蛋白与细胞凋亡[J].生物化学与生物物理学报, 2002, 34 (4): 389-394.
    [60] Bonavitaa F, Stefanellia C, Giordanoa E. H9c2 cardiac myoblasts undergo apoptosis in a model of ischemia consisting of serum deprivation and hypoxia: inhibition by PMA[J]. FEBS Lett, 2003, 536: 85-91.
    [61]O'Neill J, Manion M, Schwartz P, et al. Promises and challenges of targeting Bcl-2 anti-apoptotic proteins for cancer therapy[J]. Biochim Biophys Acta, 2004, 1705 (1): 43-51.
    [62] Skommera J, Wlodkowic D, Deptala A. Larger than life: Mitochondria and the Bcl-2 family[J]. Leukemia Res. 2007, 31: 227-286.
    [63] Misao J, Hayakawa Y, Ohno M, et al. Expression of bcl-2 protein, an inhibitor of apoptosis, and Bax, an acceleretor of apoptosis, in ventricular myocytes of human hearts with myocardial infarction[J]. Circulation, 1996, 94(7): 1506-12.
    [64]方希敏.细胞色素C与细胞凋亡[J].国外医学·临床生物化学与检验学分册, 2005, 26 (1): 43-46.
    [65] Kadenbach B, Arnold S, Lee I, et al. The possible role of cytochrome coxidase in stress-induced apoptosis and degenerative diseases[J]. Biochim Biophys Acta, 2004, 1655: 400-408.
    [66] Gogvadze V, Orrenius S, Zhivotovsky B. Multiple pathways of cytochrome c release from mitochondria in apoptosis[J]. Biochim Biophys Acta, 2006, 1757 (5-6): 146-157.
    [67] Coutant A, Lebeau J, Bidon-Wagner N, et al. Cadmium-induced apoptosis in lymphoblastoid cell line:involvement of caspase-dependent and -independent pathways[J]. Biochemstry, 2006, 88: 1815-1822.
    [68] Han W, Shi X, Nuttall AL. AIF and endoG translocation in noise exposure induced hair cell death[J]. Hearing Res, 2006, 211: 85-95.
    [69] Oda E, Ohki R, Murasawa H, et al. Noxa, a BH3-member of the bcl-2 family and candidate mediator of p53-induced apoptosis[J]. Science, 2000, 288: 1053-1058.
    [70] Leri A.Stretch-mediated release of angiotensinⅡinduces myocyte apoptosis by activating p53 that enhancea the local renin-angiotensin system and decreases the bcl-2 to bax protein ratio in the cel[J]l.J Clin Invest, 1998;101:1326-42.
    [71] Ballard-Croft C, Locklar AC, Kristo G, et al. Regional myocardial ischemia-induces activation of MAPKs is associated with subcellular redistribution of cavelin and cholesterol[J]. Am J Physiol Heart Cire Physiol, 2006; 291: H658-67.
    [72]武蓓,陈红,任景怡等.心肌缺血/再灌注损伤中细胞凋亡的信号转导通路[J].中国医药导刊, 2007; 9(5):364-7.
    [73] Li DY, Tao L, Liu H, et al. Role of ERK1/2 in the anti-apoptotic and cardioprotective effects of nitric oxide after myocardial ischemia and reperfusion[J]. Apoptosis, 2006; 11:913-30.
    [74] Janicke RU, Sprengart ML, Wati MR, et al. Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis[J]. J Biol Chem, 1998, 273: 9357-60.
    [75] Engelbrecht AM, Niesler C, Page C, et al. p38 and JNK have distinct regulatory functions on the development of apoptosis during simulated ischemia ang reperfusion in neomatal cardiomyocytes[J]. Bsdic Res Cardiol, 2004, 99(5): 338-50.
    [76] Ferrandi C, Ballerio R, Gailard P, et al. Inhibition of c-Jun N-teminal kinase decreases cardiomyocyte apoptosis and infarct size after myocardial ischemia and reperfusion in anaesthetized rats[J]. Br J Pharmacol, 2004, 142(6): 953-60.
    [77] Booz GW, Day JNE, Baker KM. Interplay between the cardiac renin angiotensin system and JAK-STAT signaling: Role in cardiac hypertrophy, ischemia/reperfusion dysfunction, and heart failure[J]. J Mol Cell Cardiol, 2002,34:1443-1453.
    [78] Mascareno E, El-Shafei M, Maulik N, et al. JAK/STAT signaling is associated with cardiac dysfunction during ischemia and reperfusion[J]. Cireculation, 2001,104:325-329.
    [79] Kisseleva T, Bhattacharya S, Braunstein J, et al. Signaling through the JAK/STAT pathway, recent advances and future challenges[J]. Gene, 2002,285:1-24.
    [80] Xuan YT, Guo Y, Han H, et al. An essential role of the JAK-STAT pathway in ischemic procongditioning[J]. Proc Natl Acad Sci USA, 2001,98:9050-9055.
    [81] Yamauchi-Takihara K, Kishimoto T. A novel fole for STAT3 in caediac remodeling[J]. Trends Cardiocasc Med, 2000,10:298-303.
    [82] Sen R, Baltimore D. Multiple muclear factors ineract with the immunoglobulin enhancer sequences [J]. Cell. 1986,46(5):705.
    [83] Hiasa G, Gamadea M, Ikeda S, et al. Ischemic preconditioning and lipopolysaccharide attenuate muclera factor-kappa B activation and gene experssion of inflammatory cytokines in the ischemis-reperfused rat heart [J]. Jpn Cire J, 2001,65(11):984-990.
    [84] Valen G.. Signal transduction through nuclear factor kappa B in ischemia- reperfusion and heart failure [J]. Basic Res Cardiol, 2004,99(1):1-7.
    [85] Morishita R, Srgimoto T, Aoki M, et al. In vivo transfection of cis element“decoy”against mucleat nuclear factor-kappa B binding site prevents myocardial infarction [J]. J Am Coll Cardiol. 2001,38(2):307-314.
    [86] Valen G, Tan ZQ, Gansson GK. Muclera factor kappa B and the heart [J]. J Am Coll Caardiol. 2001,38(2):307-314.
    [87] Fresno-Vara JA, Casado E, de-Castro J, et al. PL3K/Akt signaling pathway and cancer[J]. Cancer Treat Rev, 2004,30:193-204.
    [88] Ravingerova T, Matejikova J, Nechar J, et al. Differential role of PI3K/Akt pathway in the infarct size limitation and antiarrhythmic protection in the rat heart[J]. Mol Cell Biochem, 2006,297(1,2):111-120.
    [89] Zhu M, Feng J, Luccinetti E, et al. Ischemic postconditioning protects remodeled myocardium via the PI3K-PKB/Akt reperfusion injury salvage kinadepathway[J]. Cardiovasc Res, 2006;72:152-162.
    [90] Tskahame H, Minamino T, Huata A, et al. Gramulocyte colomy-stimuleting factor mediates cardioprotecton against ischemia/reperfusion injury via phosphatidylinositol-3-kinase/Akt pathway in canine hearts[J]. Cardiovasc Drugs Ther,2006;20:159-165.
    [91] Ye Y, Lin Y, Atar S, et al. Myocardial protection by pioglitazone, atorvastatin, and their combination: mechanisms and possible interactons[J]. Am J physiol Heart Cire Physiol Hwart Cire Physiol, 2006;291:HI158-HI169.
    [92] Rsphael J, Abedat S, Rivo J, et al. Volatile aneathetic preconditioning attenustes myocardial apoptosis in rabiea after regiona ischemia and reperfusion via Akt signaling and modulation of Bcl-2 family proteins[J]. J Pharmacol Exp Ther, 2006;318:186-194.
    [93] Mocanu MM, Bell RM,Yellon DM. PI3 kinase and not p42/p44 appears to be implicated in the protection cinferred by ischemic pwecongditioning[J]. J Mol Cell Caediol, 2002;34:661-668.
    [94] Tong H, Imashi K, Steenbergen C, et al. Phosphorylation of glycongen synthase kinase-3βduring percongditoning through a phosphatidylinositol- 3-kinase-dependent pathway is caedioprotective[J]. Cire Res, 2002;90:377-379.
    [95] Krige T, Qin Q, MeIntosh EC, et al. Ach and sdenosine activate PI3-kinase in rabbit hearts through transactivation of receptor tyrosine kinases[J]. Am J Physiol, 2002;283:H2322-H2330.
    [96] Wolfrum S, Dendorfer A, Schut tM, et al. Simvastatin acutely reduces myocardial reperfusion injury in vivo by activation the phosphatidylinositide 3-Kinade/Akt pathway[J]. J Cardiovasc Pharmacol, 2004;44:348-355.
    [97] Feng B, Stemmer PM. Ca binding site 2 in calcineurin-13 modulates calmodulin-dependent calcineurin phosphatase activity. Biochemistry[J]. 2001; 40(5):8808-8814.
    [98] Saito S, Huoi Y, Zou Y, et al.β-Adrenergic pathway induces apoptosis through calcineurin activation in cardiac myocytes [J]. J Biol Chen, 2000, 275:34528-34533.
    [99] Miyata K, Omori N, Uchino H, et al. Involvement of the brain-derived neurotuophic factor/TrkB pathway in neuroprotective effect of cyclosporin A in forebrain ischemia.[J] Neuroscience,2001, 105:571-578.
    [100] Polli JW, Billingsley ML, Kincaid RL. Expression of the calmodulin-dependent protein phosphatase, calcineurin, in rat brain: developmental patterns and the role of nigrostriatal innervation[J]. Brain Res Dev Brain Res. 1991;63(1-2):105-119.
    [101] Wallace RW, Tallant EA, Cheung WY. High levels of a heat-labile calmodulin-binding protein (CaM-BP80) in bovine neostriatum[J]. Biochemistry. 1980;19(9):1831-1837.
    [102] Giffith JP, Kim JL, Kim EE, et al. X-ray structure of Calcineurin inhibited by the immunophilin-immunosuppressant FKBP12-FK506 complex [J]. Cell, 1995, 82(3):507-522.
    [103] Wang M, Yi H,Cuerini D, et al. Calcineurin Aα, Calcineurin Aβand Calcineurin B are located on human chromosomes 4, 10q21-q22 and 2p16-p15 respectively [J]. Cytogene Cell Gene, 1996, 72(2/3):236-241.
    [104] Barford D. molecular mechanisms of the protein serine/threonine phosphatases[J]. Trends Biochem Sci, 1996, 21(11):407-412.
    [105] Lai MM, Burnett PE, Wolosker H, et al. Cain, a novel physiologic protein inhibitor of chalcineurin[J]. J Biol Chem, 1998, 273(290:18325-18331.
    [106] Crabtree GR, Clipstone NA. Sogma; tramsossopm between the plasma membrane and nucleus of T neurones[J]. J physiol. 1986;378:31-51.
    [107] Sussman MA, Lin HW, Gude N, et al. Prevention of cardiac hypertrophy in mice by calcineurin inhibition[J]. Science, 1998, 281(5383):1690-1693.
    [108] Yskel JL. Calcineurin regulation of synapic function: from ion channels to reansmitter relerse and gene transcription[J]. Trends Pharmeol Sci, 1997, 18(4):124-133.
    [109] Chen TC, Law B, Kondratyuk T, et al. Identification of soluble pwotein phosphatases that dephosphorylate voltage-sensitive sodium channels in rat brain[J]. J Biol Chem. 1995; 270(130:7750-7756.
    [110] Molkentin JD, Lu JR, Antos CL, et al. A calcineurin-dependent reanscriptiona pathway for cardiac hypertrophy. Cell, 1998, 93(2):215-228.
    [111] Lim HW, New L, Han J, et al. Calcineurine enhances MAPK phosphatase1 expression and p38 MAPK inactivarion in cardiac myocytes [J]. J Biol Chem, 2001,276(19):15913-15919.
    [112] Kakuta T, Hasegawa K, Iwai-Kanai E, er al. Calcineurin pathway isrequired for endothlin-1-mediated protection against oxidant stress-induced apoptosis in cardiac myocytes [J]. Cire Res, 2001, 88:1239-1246.
    [113] De-Windt LJ, Lim HW, Tsigen T, et al. Calcineurin mediatde hypertrophy protects cardiomyocytes from apoptosis in vitro and in vivo: an apoptosis independent midel of dilated heart failure [J]. Cire Res, 2000, 86(3):255-263.
    [114] Saito S, Hiroi Y, Zou Y, et al.β-Adrenergic pathway inducea apoptosis through calcineurin activation in cardiac myocytes [J]. J Biol Chem, 2000, 275:34528-34533.
    [115] Ranger AM, Grusby MJ, Hodge MR, et al. The transcription factor NF-Atc is essential for cardiac valve form ation[J]. Nature, 1998,392(6672):186-189.
    [116] Viola JP, Carvalho LD, Fonseca BP, et al. NFAT transcription factors: from cell cycle to tumor development[J]. Braz J Med Biol Res. 2005;38(3):335-344.
    [117] Crabtree GR, Olson EN. NFAT signaling: choreographing the social lives of cells[J].Cell. 2002; 109(Suppl):S67-S79.
    [118] Crabtree GR. The transcrption factor NFAT3 mediates neuroonal survial[J]. Cell. 1999; 96(5):611-614.
    [119] Ren S, Rollins BJ. The related retinoblastoma (pRb) and p130 proteins cooperate to regulate homeostasis in the intestinal epithelium[J]. Cell 2004; 117(2):239-251.
    [120] Wilson AJ, Jabr RI, Clapp LH. Calcium modulation of vascular smooth muscle ATP-sensitive K+ channels: role of protein phosphatase-2B[J]. Circ Res, 2000; 87(11):1019-1025.
    [121] Graef IA, Chen F, Chen L, et al. signal transduced by Ca2+/calcineurin and NFATc3/NFATc4 pattern the developing vasculature[J]. Cell, 2001;105(7):863-875.
    [122] Wang Z, Kuischke W, Richardson KE, et al. Electrical remodeling in pressure-overload cardiac hypertrophy: role of Calcineurin[J]. Circulation. 2004; 104(14):1657.
    [123] Takeda Y, Yoneda T, Demura M, et al. Calcineutin inhibition attenuates mineralocorticoid-induced cardiac hypertrophy[J]. Circulation, 2002,105(6):677-679.
    [124] Nagata K, Somuta F, Obata K, et al. AT1 receptor blochade reduces caediac calcineurin activity in hyperensive rats[J]. Hypertension, 2002, 40(2):168.
    [125]符民桂,许松,柴三葆,等.钙调神经磷酸酶依赖的信号通路在大鼠心肌肥大中的作用[J].北京医科大学学报, 1999,31:532-535.
    [126] Boss V, Abbott KL, Wang XF, et al. The cyclosporin A- sensitive nuclear factor of activated T cells (NFAT) proteins are expressed invascular smooth muscle cells[J]. J Biol Chem, 1998, 273(31):19664-49667.
    [127] Nishimura Y, Tanaka T. Calcium-dependent activation of nuclear factor regulated by interleakin3/ademovirus E4 promoter-binding peotein gene wxpression by calcineurin nuclear factor of activated T cekks and calcium/calmodulin-dependent protein kinase signaling[J]. J Biol Chem, 2001,276(23):19921-19928.
    [128] Wada H, Haswgawa K, Morimoto T, et al. Calcineurin GATA-6 pathway is involved in smooth muscle-specidic transcription[J]. J Cell Biol, 2002, 156(8): 983-991.
    [129] Sule G, Seda V, Kamil V, et al. The role of ATP sensitive K+ channels and of nitric oxide synthase on myocardial ischemia/ reperfusion-induced apoptosis[J]. Acta histochemica. 2006; 108:95-104.
    [130] Marsen TA, Simonson MS, Dunn MJ. Thrombin mediated ET-1 gene regulation involves CaM kinases and calcineurin in human endothelial cells[J]. J Cardiocasc pharmacol, 1995, 26(Suppl 3): S1-S4.
    [131]杨锐,王芬珍,吴黎明. NF-κB与心肌缺血再灌注损伤[J].心血管康复医学杂志. 2006; 15(2):193-195.
    [132] Bastiano Sanna, Orlando FB, Yan SD, et al. Direct and indirect interactions between calcineurin-NFAT and MEK1-extracellular signal-regulated kinase 1/2 signaling pathways regulate cardiac gene expression and cellular growth. Molecular and cellular biology. 2005; 25(3):865-878.
    [133] Cindy Ruwhof. Mechanical stress-induced cardiac hypertrophy: mechanisms and signal transduction pathways[J]. Cardiovascular Research.2000; 47(1):23-37.
    [134] Inmaculada OP, Eva C, Felipe W, et al. c-Jun N-terminal Kinase (JNK) Positively Regulates NFATc2 Transactivation through Phosphorylation within the N-terminal Regulatory Domain[J]. J. Biol. Chem. 2005; 280(21):20867-20878.
    [135] Qing HL, Ben JJ, WilkinsYJ, et al. Direct interaction and reciprocal regulation between ASK1 and calcineurin-NFAT control cardiomyocyte death and growth[J]. Mol Cell Biol. 2006; 26(10):3785-3797.
    [136] Jeffery DM. Calcineurin-NFAT signaling regulates the cardiac hypertrophic response in coordination with the MAPKs[J]. Cardiovascular Research.2004; 63(3):467-475.
    [137] Fanny V, Nicolas D, Christ C, et al. Dual level of interactions between calcineurin and PKC in cardiomocyte stretch[J]. Cardiovascular Research. 2006; 71(1):97-107.
    [138] Gang YW, Karl D, Richard WT. Activity-dependent CREB phosphorylation: Convergence of a fast, sensitive calmodulin kinase pathway and a slow, less sensitive mitogen-activated protein kinase psthway[J]. PNAS. 2001; 98(5):2808-2813.
    [139] Grassberger M, Steinhoff M, Schneider D, et al. Pimecrolimus: an anti-inflammatory drug targeting the skin[J]. Exp Dermatol. 2004; 13(12):721-730.
    [140] Kapturezak MH, Merer HU, Kaplan B. Pharmacolocgy of calcineurin antagonists[J]. Transplant Proc, 2004; 36(2):25S-32S.
    [141] Ke HM, Huai Q. Structures of calcineurin andits complexes with immunophilins immunosuppressants[J]. Biochem biophys Res Commun, 2003; 311(4):1095-1102.
    [142] Herzig S, Neumann J. Effects of serine/threonine protein phosphatases onion channels inexcitable membranes[J]. Physiol Rev. 2000; 80(1):173-196.
    [143] Jun OL, Luo HR, Bumett PE, et al. The calcineurin binding protein cain is a negative regulator of synaptic vesicle endocytosis[J]. Biol Chem. 2000; 275(44):34017-34020.
    [144] Sun L, Youn HD, Christine L, et al. Cabin1, an egative regulator for calcinerin signaling in Tlymphocytes[J].Immunity 1998, 8(6):703-711.
    [145] Ksng CB, Feng L, Chia J, et al. Molecular characterization of FK-506 binding protein38 and its potential regulatory role on the anti-apoptotic protein Bcl-2[J]. Biochem Biophys Res Commun. 2005; 337(1):30-38.
    [146] Weiwad M, Edlich F, Erdmann F, et al. Areass essment of the inhibitory capacity of human FKBP38 on calcineurin[J]. FEBSLett. 2005; 579:1591-1596.
    [147] Maestre MM, Edlich F, Jarczo WF, et al. Solution structure of the FK506-binding domain of human FKBP38[J]. BiomolNMR, 2006; 34(3):197-202.
    [148] Lin X, Sikkink RA, Rusnak F, et al. Inhibition of calcineurin phosphatase activity by a calcineurin phomologous protein[J]. Biol Chem, 1999; 274(51):36125-36131.
    [149] Jun OL. Endogenous protein inhibitors of calcineurin[J]. BioChemBiophys Res Commun, 2003; 311(4):1103-1109.
    [150] Dell-Acqua ML, Dodge KL, Tavalin SJ, et al. Mapping the protein phosphatase2B anchoring site on AKAP79[J]. Biol Chem, 2002; 277(50):48796-48802.

© 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