Cardioprotective Effect of Pretreatment with A Single High-Dose Atorvastatin via Regulating Myocardial Uncoupling Protein 3 in a Rat Ischemia/Reperfusion Model

2021 ◽  
Vol 11 (11) ◽  
pp. 2294-2297
Author(s):  
Yuhai Zou ◽  
Cheng Xu ◽  
Ben Zhang ◽  
Wenting Liu

Objective: Since ischemia/reperfusion (I/R) can cause malignant arrhythmia, we explored the cardioprotective effect of pretreatment with a single and large dose of atorvastatin in the SD rat model. Methods: Rats were distributed into atorvastatin (Ator), I/R model and sham groups (n = 8/group) by random number table method. In Ator group, atorvastatin was gavaged with a single dose (80 mg/kg) 12 h before I/R. The heart was treated with ischemia for 30 min and then reperfusion for 2 h. Results: Myocardial infarct area was induced by I/R when compared with Sham group. Compared with I/R group, the pretreatment of atorvastatin significantly reduced area at risk/left ventricle (40.78 ± 1.39% vs. 46.76 ± 1.42%, p < 0.01), infarct area/area at risk (21.47 ± 1.65% vs. 29.16 ± 1.21%, p < 0.01), and lactate dehydrogenase activity (3056.17 ± 136.22 RFU vs. 3864.15 ± 162.92 RFU, p < 0.05). I/R induced uncoupling protein 3 (UCP3) in transcriptional and translational levels, but atorvastatin significantly increased the UCP3 expression when compared with I/R group, 1.91 ± 0.42 vs. 1.42 ± 0.21 fold (p < 0.05) in mRNA levels measured by RT-PCR and 2.07 ± 0.18 versus 1.45 ± 0.23 fold in protein levels by Western blots. Conclusion: A single high-dose atorvastatin pretreatment 12 h before I/R reduces the infarct area in I/R model in rats. The cardioprotection may be via regulating myocardial UCP3.

2013 ◽  
Vol 113 (suppl_1) ◽  
Author(s):  
Ulrich Hofmann ◽  
Denise Mathes ◽  
Johannes Weirather ◽  
Niklas Beyersdorf ◽  
Thomas Kerkau ◽  
...  

Background: We have recently shown that CD4 + but not CD8 + T-cells contribute to ischemia-reperfusion injury of the myocardium. We therefore hypothesized that CD4 + T-cells become activated by autoantigen recognition via their T-cell receptor during reperfusion. Methods and Results: Infarct size as percent of the area-at-risk was determined by combined Evans` blue and triphenyltetrazolium (TTC) staining after 30 minutes of in vivo ischemia followed by 24 hours reperfusion in mice. After 24 hours of reperfusion there was a significantly increased population of CD4 + T-cells which expressed the surface protein CD40L in comparison to sham operated mice [n≥7; p<0.05; WT 10.8 ± 0.2% vs. sham 6.4 ± 0.5%]. CD40L is typically expressed in T-cells activated by T-cell receptor engagement. OT-II mice carry a transgenic T-cell receptor with specificity for an ovalbumin-derived peptide. These mice have a limited T-cell receptor repertoire, dominated by specificity for the irrelevant antigen ovalbumin. After 30 minutes of ischemia and 24 hours of reperfusion OT-II mice showed significantly reduction in infarct size [n≥4; p= 0.02; infarct/area at risk: OTII mice 38.9 ± 2.4% vs. WT mice 63.7 ± 6.6 % ]. Administration of a CD40L blocking antibody to wildtype mice also reduced infarct size when compared to administration of isotype-matched antibodies [n≥6; p = 0.03; infarct/ area at risk: anti-CD154 treatment 60.4 ± 3.4% vs. control 75.3 ± 4.1%]. CD4 + CD25 + Foxp3 + T-cells (natural T-regulatory cells) have a low activation threshold and constitute a T-cell subset with reactivity against autoantigens. Depletion of these cells by diphtheria-toxin application in a mouse model expressing the diphtheria-toxin receptor under the Foxp3 promotor also resulted in a significant reduction of infarct size when compared to diphtheria-toxin treated wildtype mice [n≥4; p=0.03; infarct/ area at risk: T reg -depleted DEREG mice 51.9± 3% vs. WT littermates 72.3± 2%]. Conclusion: Our results indicate that CD4 + T-cells that have been activated by an MHC class II/ T-cell receptor dependent mechanism, presumably by autoantigen recognition, contribute to myocardial ischemia-reperfusion injury.


Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Stefan Frantz ◽  
Johann Bauersachs ◽  
Anna Adamek ◽  
Nadja Blomer ◽  
Georg Ertl ◽  
...  

Formation of fibrin is critical for limiting blood loss at a site of blood vessel injury (hemostasis), but may also contribute to vascular thrombosis and thus exaggerate myocardial ischemia/reperfusion injury. Hereditary deficiency of factor XII (FXII), the protease that triggers the intrinsic pathway of coagulation in vitro, is not associated with spontaneous or excessive injury-related bleeding, but can reduce thrombus formation. Therefore, we studied the contribution of FXII for myocardial ischemia reperfusion injury. Following 30 minutes of ischemia and 24 hours of reperfusion infarct size in FXII deficient mice was significantly reduced as compared to wild type controls (infarct/area at risk, WT vs. FXII−/−, 73.5%±4.4% vs. 42.3±1.6%, p<0.001) despite similar area at risk (WT vs. FXII−/−, 28.3%±1.7% vs. 29.9±1.0%, p=n.s.). There was no difference in bleeding complications and bleeding time. Mice deficient in the FXII substrate factor XI were similarly protected from ischemia reperfusion injury (infarct/area at risk, FXI−/−, 16.9%±4.1%, vs. WT p<0.001). The phenotype of FXII−/−mice could be rescued by intravenous application of human FXII (infarct/area at risk, FXII−/−+FXII, 77.3%±8.4%, p<0.001 vs. FXIIKO). WT mice treated with the FXII inhibitor PCK (Pro-Phe-Arg-chloromethylketone) were similarly protected from myocardial ischemia/reperfusion injury (infarct/area at risk, 46.5% ±3.7%, vs. WT p<0.001). The data suggest that the intrinsic pathway is crucial for pathological clotting in myocardial ischemia reperfusion injury. FXII inhibition may offer a selective and safe strategy to reduce ischemic injury.


1992 ◽  
Vol 263 (3) ◽  
pp. H771-H777 ◽  
Author(s):  
M. R. Siegfried ◽  
C. Carey ◽  
X. L. Ma ◽  
A. M. Lefer

Intravenous administration of SPM-5185 [N-nitratopivaloyl-S-(N–-acetylalanyl)-cysteine ethyl ester], a cysteine-containing nitric oxide (NO) donor, or SPM-5267 [pivaloyl-S-(N–-acetylalanyl)-cysteine ethyl ester], an analogue of SPM-5185 that lacks the NO moiety, was studied in a feline myocardial ischemia-reperfusion model. Administration of SPM-5185 (1 mg/kg), followed by a 2-mg.kg-1.h-1 infusion starting 10 min before reperfusion, resulted in significant protection 4.5 h postreperfusion. In the myocardial ischemia (MI)+SPM-5267 group, 38 +/- 4% of the area at risk was necrotic, whereas the necrotic area/area at risk was only 7 +/- 2% in the MI+SPM-5185 group (P less than 0.01). Moreover, SPM-5185 treatment markedly attenuated the endothelial dysfunction observed in the left anterior descending coronary artery after reperfusion by 50%. These beneficial effects occurred despite the absence of a significant change in myocardial oxygen demand, as measured by the pressure-rate index. In vitro experiments demonstrated that SMP-5185, but not SPM-5267, decreased adherence of neutrophils to the coronary vascular endothelium and decreased production of superoxide radicals. Therefore, a likely mechanism of the observed cardioprotection by SPM-5185 involves attenuation of polymorphonuclear leukocyte-induced endothelial dysfunction.


2001 ◽  
Vol 226 (4) ◽  
pp. 320-327 ◽  
Author(s):  
Brian P. Lipton ◽  
Joseph B. Delcarpio ◽  
Kathleen H. McDonough

We have previously shown that a nonlethal dose of lipopolysaccharide (LPS) decreases L-selectin expression of neutrophils (PMNs), thereby preventing PMN-mediated reperfusion injury in the isolated heart. In the present study we determined whether or not that dose of LPS would protect hearts during in vivo ischemia and reperfusion by preventing PMN-induced reperfusion injury. Rats receiving saline vehicle showed marked myocardial injury (necrotic area/area at risk = 82% ± 2%) and significant depression in left ventricular function as assessed in the isolated isovolumic heart preparation at constant flow rates of 5, 10, 15, and 20 ml/min. The administration of LPS (100 μg/kg body wt) 7 hr prior to ischemia resulted in a reduction in myocardial damage (necrotic area/area at risk = 42% ± 3%) and preservation of function. Myocardial function was similar to that of sham ischemic saline- and LPS-treated rats. Moreover, PMN infiltration as determined by histology was quantitatively more severe in hearts of saline-treated rats than in hearts of LPS-treated rats. Isolated hearts from vehicle- and LPS-treated animals undergoing sham ischemia in vivo recovered to the same extent after in vitro ischemia/reperfusion, suggesting that LPS did not induce protection by altering intrinsic properties of the heart. Our results indicate that LPS-induced protection of the heart from in vivo PMN-mediated ischemia/reperfusion injury may be due to decreased L-selectin expression of PMNs in LPS-treated animals.


Author(s):  
Н.В. Нарыжная ◽  
С.Ю. Цибульников ◽  
А.В. Мухомедзянов ◽  
Л.Н. Маслов

Известно, что при хронической умеренной гипоксии формируется неспецифическая резистентность миокарда к повреждению при ишемии и следующей за ней реперфузии. Однако рецепторные механизмы формирования подобной устойчивости исследованы недостаточно. Цель исследования - изучение участия брадикининовых, каннабиноидных и ванилоидных рецепторов (TRPV1-каналов) в реализации инфаркт-лимитирующего эффекта хронической нормобарической гипоксии. Методика. Исследование выполнено на самцах крыс Вистар адаптированных к гипоксии, для чего животных подвергали непрерывной нормобарической гипоксии (ННГ) в течение 21 сут при 12% pO2, 0,3% pCO2. У крыс воспроизводили коронароокклюзию наложением лигатуры на левую нисходящую коронарную артерию в ее верхней трети на 45 мин. Реперфузию осуществляли путем освобождения лигатуры с визуальным контролем возобновления коронарного кровообращения по гиперемии ишемизированной области. Продолжительность реперфузии 2 ч. Для выявления зоны риска лигатуру вновь затягивали и в аорту вводили 5%-й раствор перманганата калия. Участок миокарда, не подвергшийся ишемии, окрашивался, неокрашенный участок являлся зоной риска. Срезы левого желудочка толщиной 2 мм окрашивали 1% раствором 2,3,5-трифенилтетразолия (37 °С, 30 мин). Размер зоны некроза и зоны риска определяли планиметрически с помощью программы Ellipse 2.02 (ViDiTo, Чешская республика). Для ингибирования каннабиноидных СВ1-рецепторов и СВ2-рецепторов использовали соответственно их селективные антагонисты римонабант (1 мг/кг) и AM630 (2,5 мг/кг); селективный антагонист HOE140 (50 мкг/кг) применяли для инактивации брадикининовых B2-рецепторов, капсазепин (3 мг/кг) - ванилоидных рецепторов (TRPV1-каналов). Все антагонисты вводили за 15 мин до коронароокклюзии. Результаты. Показано, что размер некротического повреждения миокарда у крыс адаптированных к гипоксии составляет 33% процента от размера зоны риска (53% у неадаптированных), что свидетельствует о выраженном инфаркт-лимитирующем эффекте. Этот эффект не проявлялся при ингибировании B2-брадикинировых рецепторов. Блокада каннабиноидных или ванилоидных рецепторов не влияла на инфаркт-лимитирующее действие ННГ. Следовательно, инфаркт-лимитирующий эффект ННГ зависит от активации брадикининовых B2-рецепторов, адаптационное повышение толерантности сердца к ишемии/реперфузии не зависит от каннабиноидных или ванилоидных рецепторов. Заключение. Брадикининовые рецепторы можно рассматривать в качестве одного из ключевых механизмов формирования инфаркт-лимитирующего действия ННГ. Учитывая данные о важной роли опиоидных рецепторов в кардиопротекции при ННГ, можно говорить о реализации инфаркт-лимитирующего эффекта хронической гипоксии через Gi/o-протеин-сопряженные опиоидные и брадикининовые рецепторы. Каннабиноидные рецепторы и TRPV1-каналы не участвуют в инфаркт-лимитирующем действии адаптации к нормобарической гипоксии. Aim. Chronic moderate hypoxia is known to induce nonspecific myocardial resistance to ischemia-reperfusion injury. However, receptor-mediated mechanisms of this resistance are understudied. The aim of this study was to investigate the involvement of bradykinin, cannabinoid, and vanilloid (TRPV1 channel) receptors in development of the infarction-limiting effect of chronic normobaric hypoxia (CNH). Methods. The study was performed on male Wistar rats exposed to CNH at 12% pO2 and 0.3% pCO2 for 21 days. Coronary occlusion was induced by ligation of the left descending coronary artery at the upper third of the artery for 45 min, which was followed by 2-h reperfusion produced by releasing the ligature under visual control of the recovery of coronary blood flow by hyperemia of the ischemic area. For detection of the area at risk, the ligature was tightened again, and 5% potassium permanganate solution was infused into the aorta to stain the nonischemic myocardial area. 2-mm sections of the left ventricle were stained with 1% solution of 2,3,4-triphenyl tetrazolium (37 oC, 30 min). Necrotic area and area at risk were measured planimetrically with the tEllipse 2.02 (ViDiTo, Czech Republic) software. Cannabinoid CB1 and CB2 receptors were inhibited with their respective antagonists, rimonabant (1 mg/kg) and AM630 (2.5 mg/kg); bradykinin B2 receptors were inactivated with the selective antagonist HOE140 (50 μg/kg); and vanilloid receptors (TRPV1 channels) were inhibited with capsazepine (3 mg/kg). All antagonists were administered 15 minutes prior to coronary occlusion. Results. The size of necrotic area was 33% of the area at risk in rats adapted to hypoxia vs. 53% in non-adapted rats. This infarct-limiting effect of adaptation to hypoxia was abolished by inhibition of B2-bradykinin receptors. Blockade of cannabinoid or vanilloid receptors did not change the infarct-limiting effect of CNH. Therefore, the infarction-limiting effect of CNH depends on activation of bradykinin B2 receptors but not of cannabinoid or vanilloid receptors. Conclusion. Activation of bradykinin receptors can be considered a key mechanism of the infarct-limiting effect of CNH. Since opioid receptors are known to play an important role in CNH cardioprotection, the infarct-limiting effect of CNH may be mediated by Gi/o-coupled opioid and bradykinin receptors. Cannabinoid receptors and TRV1-channels do not contribute to the infarct-limiting effect of adaptation to normobaric hypoxia.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 771-771
Author(s):  
Li Zhu ◽  
Tao Wang ◽  
Hong Jiang ◽  
Timothy J. Stalker ◽  
Karen P. Fong ◽  
...  

Abstract Abstract 771 Introduction. Platelets are activated early during the reperfusion of ischemic myocardium, potentially exacerbating the extent of ischemia/reperfusion (I/R) injury. We have recently shown (Zhu, et al., PNAS 2007) that platelets express the semaphorin family member, sema4D, as do T-cells. Sema4D is a cell surface protein whose receptors are expressed by B-cells, monocytes and endothelial cells as well as platelets. Loss of sema4D expression in mice causes a defect in signaling downstream of the platelet collagen receptor, glycoprotein (GP) VI, inhibiting platelet function in vitro and in vivo, and reducing the extent of platelet hyperresponsiveness and atherothrombosis in the setting of dyslipidemia (Zhu, et al. ATVB 2009). Because of the role played by platelets, leukocytes and endothelial cells in reperfusion injury, here we asked whether loss of sema4D expression can also protect the heart, reducing the extent of damage following temporary ischemia. Methods. The left anterior descending coronary artery of anaesthetized mice was ligated for 45 min. After reperfusion for 48 hours, the mice were re-anesthetized and perfused with 2,3,5-triphenyltetrazolium chloride to measure the area of infarction. Fluorescent microspheres were used to delineate the area at risk. Comparisons were made between sema4D(−/−) and wild type mice produced by breeding sema4D(+/−) heterozygotes. Results. Although there was no difference between the sema4D(−/−) and WT mice with respect to either heart size or area at risk, we observed a substantial (57%) decrease in infarct size in the sema4D(−/−) mice expressed as a fraction of the area at risk (N=7-9, p<0.005). Since sema4D is shed from the surface of activated platelets and T-cells by the metalloprotease, ADAM17, producing a large bioactive fragment, we next asked whether the protection against ischemia/reperfusion injury conferred by the sema4D knockout is due to the loss of cell-associated or soluble sema4D. Chimeric mice were produced in which hematopoiesis was reconstituted in irradiated sema4D(+/+) mice using fetal liver cells from mouse embryos that lack functional ADAM17. This produces mice in which sema4D is expressed as usual in the hematopoietic lineages, but unable to be shed. Chimerism, inhibition of sema4D shedding and recovery of normal cell counts were confirmed after transplantation. The ischemia/reperfusion studies were repeated comparing chimeras reconstituted with ADAM17-deficient and ADAM17-replete fetal liver cells. In contrast to the sema4D knockout, the extent of infarction was the same whether or not ADAM17 was functional and sema4D was shed. Conclusions. Although the role of sema4D and its receptors have been studied most extensively in the context of T-cell interactions with B-cells, our previous studies have made a case for the involvement of sema4D in platelet:platelet and platelet:endothelial cell interactions. We now show for the first time that 1) loss of sema4D expression in mice confers protection against ischemia/reperfusion injury in the myocardium, and 2) preventing the formation of soluble, bioactive sema4D is insufficient to recapitulate this effect. Since sema4D and its receptors are expressed on more than just platelets, it cannot be concluded that the observed protection in the knockout is solely due to the absence of platelet sema4D. However, experience with other knockouts that reduce platelet function suggests that the defects that we have observed in sema4D(−/−) platelet function are likely to contribute. Regardless of whether expression on platelets is entirely or only partly responsible for the observed phenotype, sema4D is an interesting target for therapeutic intervention. Disclosures: No relevant conflicts of interest to declare.


2012 ◽  
Vol 303 (10) ◽  
pp. H1229-H1236 ◽  
Author(s):  
Gabriella F. Kocsis ◽  
Márta Sárközy ◽  
Péter Bencsik ◽  
Márton Pipicz ◽  
Zoltán V. Varga ◽  
...  

Metabolic diseases such as hyperlipidemia and diabetes attenuate the cardioprotective effect of ischemic preconditioning. In the present study, we examined whether another metabolic disease, prolonged uremia, affects ischemia/reperfusion injury and cardioprotection by ischemic preconditioning. Uremia was induced by partial nephrectomy in male Wistar rats. The development of uremia was verified 29 wk after surgery. Transthoracic echocardiography was performed to monitor cardiac function. At week 30, hearts of nephrectomized and sham-operated rats were isolated and subjected to a 30-min coronary occlusion followed by 120 min reperfusion with or without preceding preconditioning induced by three intermittent cycles of brief ischemia and reperfusion. In nephrectomized rats, plasma uric acid, carbamide, and creatinine as well as urine protein levels were increased as compared with sham-operated controls. Systolic anterior and septal wall thicknesses were increased in nephrectomized rats, suggesting the development of a minimal cardiac hypertrophy. Ejection fraction was decreased and isovolumic relaxation time was shortened in nephrectomized rats demonstrating a mild systolic and diastolic dysfunction. Infarct size was not affected significantly by nephrectomy itself. Ischemic preconditioning significantly decreased infarct size from 24.8 ± 5.2% to 6.6 ± 1.3% in the sham-operated group and also in the uremic group from 35.4 ± 9.5% to 11.9 ± 3.1% of the area at risk. Plasma ANG II and nitrotyrosine were significantly increased in the uremic rats. We conclude that although prolonged experimental uremia leads to severe metabolic changes and the development of a mild myocardial dysfunction, the cardioprotective effect of ischemic preconditioning is still preserved.


2016 ◽  
Vol 94 (9) ◽  
pp. 973-978 ◽  
Author(s):  
Li-Min Meng ◽  
Hui-Jie Ma ◽  
Hui Guo ◽  
Qian-Qian Kong ◽  
Yi Zhang

Naringenin (Nari) has antioxidative and anti-atherosclerosis effects, and activation of ATP-sensitive potassium channel (KATP) can offer cardiac protection. We hypothesized that Nari protects the heart against ischemia–reperfusion (I–R) injury through activation of KATP. Isolated hearts from adult male Sprague-Dawley rats experienced a 30-min global ischemia followed by 60-min reperfusion (120 min for the infarct size determination). The hearts were treated with Nari (NARI); Nari plus glibenclamide (GLI), a non-specific ATP-sensitive potassium channel blocker (NARI+GLI); and Nari plus 5-hydroxy decanoic acid (5-HD), a mitochondrial membrane ATP-sensitive potassium channel blocker (NARI+5-HD). The left ventricular pressure, lactate dehydrogenates (LDH) in coronary effluent, superoxide dismutase (SOD) and malondialdehyde (MDA) in myocardium, and myocardial infarct area were measured. Nari above 2.5 μmol/L improved the recovery of left ventricular function, decreased LDH in coronary effluent, and reduced myocardial infarct area. The SOD activity was increased and MDA was decreased in Nari-treated myocardium. The cardioprotective effect of Nari was canceled by GLI and 5-HD. In conclusion, Nari has a cardioprotective effect against I–R injury, which may be carried out through activating ATP-sensitive potassium channels in both cell and mitochondrial membrane, and enhancing myocardial antioxidant capacity.


2004 ◽  
Vol 17 (9) ◽  
pp. 948-953 ◽  
Author(s):  
Ana Clara Tude Rodrigues ◽  
Ryuji Hataishi ◽  
Fumito Ichinose ◽  
Kenneth D. Bloch ◽  
Genevieve Derumeaux ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document