首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 187 毫秒
1.
肾神经在肾缺血预处理对麻醉家兔心脏保护中的作用   总被引:14,自引:3,他引:11  
Ding YF  Zhang MM  He RR 《生理学报》2001,53(1):7-12
在氨基甲酸乙酯麻醉家兔上,观察肾脏缺血预处理(RIP)对缺血-再灌注心肌的影响,旨在证实RIP对心肌有无保护效应,并明确肾神经在其中的作用。所得结果如下(1)在心脏45min缺血和180min再灌注过程中,血压、心率和心肌耗氧量呈进行性下降;心外膜电图ST段在缺血期明显抬高,再灌注过程中逐渐恢复到基础对照值。心肌梗塞范围占缺血心肌的55.80±1.25%。(2)RIP时心肌梗塞范围为36.51±2.8%,较单纯心肌缺血-再灌注显著减少(P<0.01),表明RIP对心肌有保护作用。(3)肾神经切断可取消RIP对心肌的保护效应,但肾神经切断本身对单纯缺血-再灌注所致的心肌梗死范围无明显影响。(4)肾缺血(10min)时,肾传入神经放电活动由0.14±0.08增至0.65±0.12imp/s(P<0.01)。(5)预先应用腺苷受体拮抗剂8-苯茶碱可明显减弱肾缺血所激活的肾传入神经活动,提示肾传入活动的增强是由肾缺血产生的腺苷所介导。以上结果表明,肾短暂缺血-再灌注所诱发的肾神经传入活动在RIP心肌保护效应中起重要作用。  相似文献   

2.
Dong JH  Liu YX  Zhao J  Ma HJ  Guo SM  He RR 《生理学报》2004,56(5):620-624
通过氨基甲酸乙酯麻醉大鼠观察股神经电刺激对缺血- 再灌注心肌的影响,旨在证实外周神经刺激对心肌有无保护效应,并明确其可能的作用机制。心肌缺血区和梗塞区分别用伊文思蓝和氯化硝基四氮唑蓝染色确定,心肌梗塞范围定义为心肌梗塞区重量占心肌缺血区重量的百分比。所得结果如下:(1)在心肌缺血30 min 和再灌注120 min 过程中,梗塞心肌占缺血心肌的(54.96±0.82)%。 高频电刺激(10 V,100 Hz,1ms)股神经5 min 可使心肌梗塞范围减少到(36.94±1.34)% (P<0.01), 表明 (2)高频电刺激股神经对缺血-再灌注心肌有保护作用。然而,低频电刺激(10 V, 10 Hz, 1 ms)股神经对缺血-再灌注心肌梗塞范围无影响。 预先应用非选择性阿片肽受体阻断剂纳洛酮(5 mg/kg, i.v.)或非选择性KATP 通道阻断剂格列苯脲(5 mg/kg, i.v.)均能完全 (3)取消高频电刺激股神经对缺血-再灌注心肌的保护作用。以上结果提示,高频外周神经刺激可以减小缺血- 再灌注心肌的梗塞范围,其可能的作用机制是: 高频电刺激股神经时中枢神经系统内释放的内源性阿片肽和由此激活的心肌KATP通道的开放介导了这种保护作用。  相似文献   

3.
目的:观察骨骼肌缺血后处理(RPostC)、心肌的缺血后处理(MPostC)对缺血/再灌注心肌保护作用是否存在差异以及两者联合后作用是否叠加。方法:健康新西兰大白兔3O只,随机分为5组(n=6):缺血对照组(Con)、假手术组(sham)、心肌缺血后处理组(MPostC)和肢体缺血后处理组(RPostC)及心肌缺血后处理联合肢体缺血后处理组(MPostC+RPostC)。采用开胸结扎冠状动脉左室支45 min,再灌注120min方法制作缺血/再灌注模型,采用短暂结扎双侧髂外动脉固定部位5 min造成骨骼肌短暂缺血。以Evans蓝标记心肌缺血区范围,以TTC法检测梗死心肌范围,并分别于缺血前、后及再灌注1、2 h测定血浆磷酸肌酸激酶(CPK)活性和乳酸脱氢酶(LDH)含量。结果:和Con组相比,MPostC和RPostC组心肌梗死范围均明显降低(P<0.05);MPostC联合RPostC组心肌梗死范围与MPostC或RPostC组相比,均进一步降低(均P<0.05)。但MPostC组及RPostC组之间心肌坏死范围未见统计学差异。再灌注120 min末血浆CPK活性及LDH含量也显示相似趋势。结论:骨骼肌缺血后处理及心肌后处理对缺血/再灌注心肌均具有明显保护作用;且两者作用可以叠加;但骨骼肌和心肌后处理之间保护作用未显示统计学差异。  相似文献   

4.
人参皂甙 Rb1与Re对大鼠缺血再灌注心肌细胞凋亡的影响   总被引:15,自引:0,他引:15  
目的观察人参皂甙Rb1与Re对缺血再灌注心肌细胞凋亡的影响,并比较两者的效应差异.方法结扎Wistar大鼠左冠状动脉前降支,建立大鼠缺血再灌注动物模型;采用透射电镜、缺口末端标记法检测心肌凋亡细胞,利用光学显微镜进行细胞计数.结果 (1)透射电镜发现缺血再灌注组缺血区出现心肌凋亡细胞,假手术组未发现心肌凋亡细胞;(2)缺血再灌注组心肌细胞凋亡数为134.45±45.61个/视野,人参皂甙Rb1治疗组51.65±13.71个/视野,人参皂甙Re治疗组90.66±19.22个/视野,三组间有非常显著性差异(P<0.01).结论心肌缺血再灌注诱导心肌细胞凋亡,人参皂甙Rb1和Re均可显著减少缺血再灌注心肌细胞的凋亡.证实人参皂甙Rb1与Re均有抑制缺血再灌注心肌细胞凋亡,减轻心肌缺血再灌注损伤的作用;人参皂甙Rb1的抗心肌细胞凋亡作用较Re的效果为佳.  相似文献   

5.
目的研究芹菜素对缺血再灌注大鼠心肌核转录因子-κB(NF-κB)和粘附分子ICAM-1表达的影响,探讨其抗心肌缺血再灌注损伤的可能机制。方法将Wistar大鼠随机分为6组:假手术组、缺血再灌注组(IR组)、溶剂对照组、维拉帕米阳性对照组、芹菜素低、高剂量用药组。采用结扎左冠状动脉前降支制作缺血再灌注模型,心肌缺血30 min,再灌注2 h。免疫组化染色检测心肌组织的NF-κB和ICAM-1的表达.结果芹菜素组可降低心肌组织NF-κB和ICAM-1的表达,与IR组比较有显著差异(P0.05)。结论芹菜素对缺血再灌注心肌的保护作用可能与其减少心肌缺血再灌注后NF-κB和ICAM-的激活有关。  相似文献   

6.
Zhao HG  Li WB  Li QJ  Chen XL  Liu HQ  Feng RF  Ai J 《生理学报》2004,56(3):407-412
探探讨肢体缺血预处理(limb ischemic preconditioning,LIP)对大鼠全脑缺血再灌注后海马CA1区锥体细胞凋亡的影响。46只大鼠椎动脉凝闭后分为假手术组、肢体缺血组、脑缺血组、LIP组。重复夹闭大鼠双侧股动脉3次(每次10min,间隔10min)作为LIP,之后立即夹闭双侧颈总动脉进行全脑缺血8min后再灌注。DNA凝胶电泳、TUNEL和吖啶橙/溴乙锭(AO/EB)双染技术从生化和形态学方面观察海马神经元凋亡的情况。凝胶电泳显示,脑缺血组出现了凋亡特征性DNA梯状条带,而LIP组无上述条带出现。与脑缺血组比较,LIP可明显减少海马CAI区TUNEL阳性神经元数(17.8±5.8vs 69.8±12,P<0.01)。AO/EB染色也显示LIP可明显减少脑缺血再灌注引起的神经元凋亡。以上结果提示,LIP可抑制脑缺血再灌注后海马神经元的凋亡,进而减轻脑缺血再灌注损伤,提供脑保护作用。  相似文献   

7.
目的:近期实验研究显示,在再灌注的早期给予短暂、重复的缺血再灌(缺血后处理Postconditioning)能够减轻心肌再灌注损伤。本实验旨在探明三磷酸腺苷(ATP)用于缺血后处理是否产生上述保护效应,以及了解腺苷受体在此保护作用机制中的地位。方法:家兔开胸后左前降支均给予40min结扎和180min的再灌注,并随机分为5组:(1)对照组;(2)缺血后处理组;(3)ATP后处理组;(4)缺血后处理 SPT(硫苯茶碱)组;(5)SPT对照组。于实验终点测定心肌梗死面积(TTC染色),血浆CK-MB、SOD、MDA含量。结果:和时照组相比,缺血后处理组与ATP后处理组心梗面积减少(p<0.05),CK-MB也显著降低(p相似文献   

8.
肢体缺血预处理减轻大鼠海马缺血/再灌注损伤   总被引:10,自引:0,他引:10  
目的:探讨肢体缺血预处理(LIP)对大鼠全脑缺血/再灌注损伤的影响.方法: 36只大鼠椎动脉凝闭后随机分为假手术(Control)组、脑缺血组、肢体缺血组、LIP 0 d组(LIP后即刻行脑缺血)、LIP 1 d组(LIP后1 d行脑缺血)和LIP 2 d组(LIP后2 d行脑缺血).重复夹闭大鼠双侧股动脉3次(每次10 min,间隔10 min)作为LIP,夹闭颈总动脉进行全脑缺血8 min后再灌注.硫堇染色观察海马CA1区组织学分级及锥体神经元密度以判断海马损伤程度.结果:脑缺血组海马CA1区锥体神经元损伤严重,与Control组比较,组织学分级明显升高,神经元密度明显降低(P<0.01).LIP 0 d组海马CA1区神经元损伤较脑缺血组明显减轻,组织学分级明显降低,神经元密度明显升高(P<0.01).而LIP 1 d组和LIP 2 d组大鼠海马CA1区锥体细胞缺失较多,仍有明显的组织损伤.结论:LIP可减轻随后立即发生的脑缺血/再灌注损伤,但对间隔1 d后的脑缺血/再灌注损伤无显著对抗作用.  相似文献   

9.
尾加压素Ⅱ对正常及缺血-再灌注离体大鼠心脏的影响   总被引:6,自引:1,他引:5  
Zhou P  Wu SY  Yu CF  Wang H  Tang CS  Lin L  Yuan WJ 《生理学报》2003,55(4):442-448
在正常Langendorff灌流与缺血-再灌注(停灌20 min-复灌20 min)离体大鼠心脏模型,观察尾加压素Ⅱ(urotensin Ⅱ,UⅡ)对冠脉流量、心功能和心肌代谢的影响以及心肌UⅡ受体的功能,以探讨UⅡ的心脏效应。对正常心脏给予0.1、1和10 nmol/L UⅡ各5 min,然后换洗5 min,对停灌缺血-再灌注心脏在再灌注期给予1或10nmol/L UⅡ。监测心率、左室内压和左室内压升降的最大变化率等心功能指标,计算冠脉流量,测定冠脉流出液中总蛋白和肌红蛋白含量以及乳酸脱氢酶(LDH)活性。灌流结束后,测定心肌丙二醛(MDA)含量和质膜UⅡ结合位点(放射性配基结合法)。结果如下:(1)正常心脏灌流UⅡ后,冠脉流量和心功能呈浓度依赖下降,换洗后没有完全恢复。心肌蛋白、肌红蛋白和LDH漏出随UⅡ浓度的增加而增加,换洗后迅速减少。UⅡ组心肌MDA含量与对照组差异无显著性。(2)缺血-再灌注后,冠脉流量显著减少,心功能显著抑制,再灌注期心肌蛋白、肌红蛋白和LDH明显漏出;给予UⅡ后,上述变化增强,且高浓度组更强,与对照组差异有显著性(P<<0.01),再灌注后心肌MDA含量亦显著高于对照(P<0.01)。(3)缺血-再灌注心肌质膜UⅡ受体的B_(max)显著高于正常对照心肌(14.65±1.78vs20.53±1.98 fmol/mg pr,P<0.01),Kd值变化无统计学意义。上述结果表明,在正常  相似文献   

10.
目的:探讨非创伤性肢体缺血预处理对缺血/再灌注心肌的作用及核因子kappa B(NF-kB)在诱导远隔器官预处理中的可能机制.方法:Wistar大鼠48只,制备心肌缺血/再灌注模型,随机分3组,缺血/再灌注组(I/R组),非创伤性肢体缺血预处理组(PL组),ProDTC非创伤性肢体缺血预处理组(PL-D组).观察各组心肌梗死面积,并应用反转录PCR技术,测定心肌组织NF-kB mRNA.结果:心肌梗死面积PL组较I/R组明显减少,分别为34.5%±7.6%和58.5%±8.5%(P<0.05),而PL-D组与I/R组相比无显著差异.与I/R组比较,PL组和PL-D组NF-kB mRNA表达明显减弱(P<0.05);PL-D组NF-kB mRNA表达较PL组亦明显减弱(P<0.05).结论:非创伤性肢体缺血预处理对缺血再灌注心肌有早期保护作用,NF-kB在肢体缺血预处理的早期心肌保护中起重要作用.  相似文献   

11.
This study investigated whether aldose reductase (AR) inhibition with zopolrestat, either alone or in combination with an adenosine A(3)-receptor agonist (CB-MECA), reduced myocardial ischemic injury in rabbit hearts subjected to 30 min of regional ischemia and 120 min of reperfusion. Zopolrestat reduced infarct size by up to 61%, both in vitro (2 nM to 1 microM; EC(50) = 24 nM) and in vivo (50 mg/kg). Zopolrestat reduced myocardial sorbitol concentration (index of AR activity) by >50% (control, 15.0 +/- 2.2 nmol/g; 200 nM zopolrestat, 6.7 +/- 1.3 nmol/g). A modestly cardioprotective concentration of CB-MECA (0.2 nM) allowed a 50-fold reduction in zopolrestat concentration while providing a similar reduction in infarct size (infarct area/area at risk: control, 62 +/- 2%; 1 microM zopolrestat, 24 +/- 5%; 20 nM zopolrestat plus 0.2 nM CB-MECA, 20 +/- 4%). In conclusion, AR inhibition is cardioprotective both in vitro and in vivo. Furthermore, combining zopolrestat with an A(3) agonist allows a reduction in the zopolrestat concentration while maintaining an equivalent degree of cardioprotection.  相似文献   

12.
The relative roles of free-radical production, mitochondrial ATP-sensitive K+ (mitoKATP) channels and possible receptor cross-talk in both opioid and adenosine A1 receptor (A1AR) mediated protection were assessed in a rat model of myocardial infarction. Sprague-Dawley rats were subjected to 30 min of occlusion and 90 min of reperfusion. The untreated rats exhibited an infarct of 58.8 +/- 2.9% [infarct size (IS)/area at risk (AAR), %] at the end of reperfusion. Pretreatment with either the nonselective opioid receptor agonist morphine or the selective A1AR agonist 2-chloro-cyclopentyladenosine (CCPA) dramatically reduced IS/AAR to 41.1 +/- 2.2% and 37.9 +/- 5.5%, respectively (P < 0.05). Protection afforded by either morphine or CCPA was abolished by the reactive oxygen species scavenger N-(2-mercaptopropionyl)glycine or the mitoKATP channel blocker 5-hydroxydecanoate. Both morphine- and CCPA-mediated protection were attenuated by the selective A1AR antagonist 1,3-dipropyl-8-cyclopentylxanthine and the selective delta1-opioid receptor (DOR) antagonist 7-benzylidenealtrexone. Simultaneous administration of morphine and CCPA failed to enhance the infarct-sparing effect of either agonist alone. These data suggest that both DOR and A1AR-mediated cardioprotection are mitoKATP and reactive oxygen species dependent. Furthermore, these data suggest that there are converging pathways and/or receptor cross-talk between A1AR- and DOR-mediated cardioprotection.  相似文献   

13.
The aim of this study was to determine whether erythropoietin (EPO) affords additional cardioprotection to the preconditioned myocardium by enhanced phosphorylation of Akt, STAT3, or glycogen synthase kinase-3beta (GSK-3 beta). Preconditioning (PC) with 5-min ischemia/5-min reperfusion and EPO (5,000 U/kg iv) reduced infarct size (as % of area at risk, %IS/AR) after 20-min ischemia in rat hearts in situ from 56.5 +/- 1.8% to 25.2 +/- 2.1% and to 36.2 +/- 2.8%, respectively. PC-induced protection was significantly inhibited by a protein kinase C inhibitor, chelerythrine (5 mg/kg), and slightly blunted by a phosphatidylinositol-3-kinase inhibitor, wortmannin (15 microg/kg). The opposite pattern of inhibition was observed for EPO-induced protection. The combination of PC and EPO further reduced %IS/AR to 8.9 +/- 1.9%, and this protection was inhibited by chelerythrine and wortmannin. The additive effects of PC and EPO on infarct size were mirrored by their effects on the level of phosphorylated GSK-3 beta at 5 min after reperfusion but not their effects on the level of phospho-Akt or phospho-STAT3. To mimic phosphorylation-induced inhibition of GSK-3 beta activity, SB-216763 (SB), a GSK-3 beta inhibitor, was administered before ischemia or 5 min before reperfusion. Infarct size was significantly reduced by preischemic injection (%IS/AR = 40.4 +/- 2.2% by 0.6 mg/kg SB and 34.0 +/- 1.8% by 1.2 mg/kg SB) and also by prereperfusion injection (%IS/AR = 32.0 +/- 2.0% by 1.2 mg/kg SB). These results suggest that EPO and PC afford additive infarct size-limiting effects by additive phosphorylation of GSK-3beta at the time of reperfusion by Akt-dependent and -independent mechanisms.  相似文献   

14.
Hydroxymethyl glutaryl (HMG)-coenzyme A (CoA) reductase inhibitors (statins) protect the myocardium against ischemia-reperfusion injury via a mechanism unrelated to cholesterol lowering. Statins may inhibit isoprenylation and thereby prevent activation of proteins such as RhoA. We hypothesized that statins protect the myocardium against ischemia-reperfusion injury via a mechanism involving inhibition of geranylgeranyl pyrophosphate synthesis and translocation of RhoA to the plasma membrane. Sprague-Dawley rats were given either the HMG-CoA reductase inhibitor rosuvastatin, geranylgeranyl pyrophosphate dissolved in methanol, the combination of rosuvastatin and geranylgeranyl pyrophosphate, rosuvastatin and methanol, or distilled water (control) by intraperitoneal injection for 48 h before ischemia-reperfusion. Animals were anesthetized and either subjected to 30 min of coronary artery occlusion followed by 2 h of reperfusion where at infarct size was determined, or the expression of RhoA protein was determined in cytosolic and membrane fractions of nonischemic myocardium. There were no significant differences in hemodynamics between the control group and the other groups before ischemia or during ischemia and reperfusion. The infarct size was 80 +/- 3% of the area at risk in the control group. Rosuvastatin reduced infarct size to 64 +/- 2% (P<0.001 vs. control). Addition of geranylgeranyl pyrophosphate (77 +/- 2%, P<0.01 vs. rosuvastatin) but not methanol (65 +/- 2%, not significant vs. rosuvastatin) abolished the cardioprotective effect of rosuvastatin. Geranylgeranyl pyrophosphate alone did not affect infarct size per se (84 +/- 2%). Rosuvastatin increased the cytosol-to-membrane ratio of RhoA protein in the myocardium (P<0.05 vs. control). These changes were abolished by addition of geranylgeranyl pyrophosphate. We conclude that the cardioprotection and the increase of the RhoA cytosol-to-membrane ratio induced by rosuvastatin in vivo are blocked by geranylgeranyl pyrophosphate. The inhibition of geranylgeranyl pyrophosphate formation and subsequent modulation of cytosol/membrane-bound RhoA are of importance for the protective effect of statins against myocardial ischemia-reperfusion injury.  相似文献   

15.
There is increasing evidence for interactions among adenosine receptor subtypes in the brain and heart. The purpose of this study was to determine whether the adenosine A(2a) receptor modulates the infarct size-reducing effect of preischemic administration of adenosine receptor agonists in intact rat myocardium. Adult male rats were submitted to in vivo regional myocardial ischemia (25 min) and 2 h reperfusion. Vehicle-treated rats were compared with rats pretreated with the A(1) agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA, 10 mug/kg), the nonselective agonist 5'-N-ethylcarboxamidoadenosine (NECA, 10 mug/kg), or the A(2a) agonist 2-[4-(2-carboxyethyl)phenethylamino]-5'-N-methylcarboxamidoadenosine (CGS-21680, 20 mug/kg). Additional CCPA- and NECA-treated rats were pretreated with the A(1) antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 100 mug/kg), the A(2a)/A(2b) antagonist 4-(-2-[7-amino-2-{2-furyl}{1,2,4}triazolo{2,3-a} {1,3,5}triazin-5-yl-amino]ethyl)phenol (ZM-241385, 1.5 mg/kg) or the A(3) antagonist 3-propyl-6-ethyl-5[(ethylthio)carbonyl]-2-phenyl-4-propyl-3-pyridine carboxylate (MRS-1523, 2 mg/kg). CCPA and NECA reduced myocardial infarct size by 50% and 35%, respectively, versus vehicle, but CGS-21680 had no effect. DPCPX blunted the bradycardia associated with CCPA and NECA, whereas ZM-241385 attenuated their hypotensive effects. Both DPCPX and ZM-241385 blocked the protective effects of CCPA and NECA. The A(3) antagonist did not alter the hemodynamic effects of CCPA or NECA, nor did it alter adenosine agonist cardioprotection. None of the antagonists alone altered myocardial infarct size. These findings suggest that although preischemic administration of an A(2a) receptor agonist does not induce cardioprotection, antagonism of the A(2a) and/or the A(2b) receptor blocks the cardioprotection associated with adenosine agonist pretreatment.  相似文献   

16.
The role of A(1) adenosine receptors (A(1)AR) in ischemic preconditioning was investigated in isolated crystalloid-perfused wild-type and transgenic mouse hearts with increased A(1)AR. The effect of preconditioning on postischemic myocardial function, lactate dehydrogenase (LDH) release, and infarct size was examined. Functional recovery was greater in transgenic versus wild-type hearts (44.8 +/- 3.4% baseline vs. 25.6 +/- 1.7%). Preconditioning improved functional recovery in wild-type hearts from 25.6 +/- 1.7% to 37.4 +/- 2.2% but did not change recovery in transgenic hearts (44.8 +/- 3.4% vs. 44.5 +/- 3.9%). In isovolumically contracting hearts, pretreatment with selective A(1) receptor antagonist 1, 3-dipropyl-8-cyclopentylxanthine attenuated the improved functional recovery in both wild-type preconditioned (74.2 +/- 7.3% baseline rate of pressure development over time untreated vs. 29.7 +/- 7.3% treated) and transgenic hearts (84.1 +/- 12.8% untreated vs. 42.1 +/- 6.8% treated). Preconditioning wild-type hearts reduced LDH release (from 7,012 +/- 1,451 to 1,691 +/- 1,256 U. l(-1). g(-1). min(-1)) and infarct size (from 62.6 +/- 5.1% to 32.3 +/- 11.5%). Preconditioning did not affect LDH release or infarct size in hearts overexpressing A(1)AR. Compared with wild-type hearts, A(1)AR overexpression markedly reduced LDH release (from 7,012 +/- 1,451 to 917 +/- 1,123 U. l(-1). g(-1). min(-1)) and infarct size (from 62.6 +/- 5.1% to 6.5 +/- 2.1%). These data demonstrate that murine preconditioning involves endogenous activation of A(1)AR. The beneficial effects of preconditioning and A(1)AR overexpression are not additive. Taken with the observation that A(1)AR blockade equally eliminates the functional protection resulting from both preconditioning and transgenic A(1)AR overexpression, we conclude that the two interventions affect cardioprotection via common mechanisms or pathways.  相似文献   

17.
We investigated the role of p38 mitogen-activated protein kinase (MAPK) phosphorylation and opening of the mitochondrial ATP-sensitive K(+) [(K(ATP))(mito)] channel in the adenosine A(1) receptor (A(1)AR)-induced delayed cardioprotective effect in the mouse heart. Adult male mice were treated with vehicle (5% DMSO) or the A(1)AR agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA; 0.1 mg/kg ip). Twenty-four hours later, hearts were subjected to 30 min of global ischemia and 30 min of reperfusion in the Langendorff mode. Genistein or SB-203580 (1 mg/kg i.p.) given 30 min before CCPA treatment was used to block receptor tyrosine kinase or p38 MAPK phosphorylation, respectively. 5-Hydroxydecanoate (5-HD; 200 microM) was used to block (K(ATP))(mito) channels. CCPA produced marked improvement in left ventricular function, which was partially blocked by SB-203580 and 5-HD and completely abolished with genistein. CCPA caused a reduction in infarct size (12.0 +/- 2.0 vs. 30.3 +/- 3.0% in vehicle), which was blocked by genistein (29.4 +/- 2.3%), SB-203580 (28.3 +/- 2.6%), and 5-HD (33.9 +/- 2.4%). CCPA treatment also caused increased phosphorylation of p38 MAPK during ischemia, which was blocked by genistein, SB-203580, and 5-HD. The results suggest that A(1)AR-triggered delayed cardioprotection is mediated by p38 MAPK phosphorylation. Blockade of cardioprotection with 5-HD concomitant with decrease in p38 MAPK phosphorylation suggests a potential role of (K(ATP))(mito) channel opening in phosphorylation and ensuing the late preconditioning effect of A(1)AR.  相似文献   

18.
The mechanism underlying interorgan preconditioning of the heart remains elusive, although a role for adenosine and activation of a neurogenic pathway has been postulated. We tested in rats the hypothesis that adenosine released by the remote ischemic organ stimulates local afferent nerves, which leads to activation of myocardial adenosine receptors. Preconditioning with a 15-min mesenteric artery occlusion (MAO15) reduced infarct size produced by a 60-min coronary artery occlusion (60-min CAO) from 68 +/- 2% to 48 +/- 4% (P < 0.05). Pretreatment with the ganglion blocker hexamethonium or 8-(p-sulfophenyl)theophylline (8-SPT) abolished the protection by MAO15. Intramesenteric artery (but not intraportal vein) infusion of adenosine (10 microg/min) was as cardioprotective as MAO15, which was also abolished by hexamethonium. Whereas administration of hexamethonium at 5 min of reperfusion following MAO15 had no effect, 8-SPT at 5 min of reperfusion abolished the protection. Permanent reocclusion of the mesenteric artery before the 60-min CAO enhanced the cardioprotection by MAO15 (30 +/- 5%), but all protection was abolished when 8-SPT was administered after reocclusion of the mesenteric artery. Together, these findings demonstrate the involvement of myocardial adenosine receptors. We therefore conclude that locally released adenosine during small intestinal ischemia stimulates afferent nerves in the mesenteric bed during early reperfusion, initiating a neurogenic pathway that leads to activation of myocardial adenosine receptors.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号