Obestatin affords cardioprotection to the ischemic-reperfused isolated rat heart and inhibits apoptosis in cultures of similarly stressed cardiomyocytes

2010 ◽  
Vol 299 (2) ◽  
pp. H470-H481 ◽  
Author(s):  
Giuseppe Alloatti ◽  
Elisa Arnoletti ◽  
Eleonora Bassino ◽  
Claudia Penna ◽  
Maria Giulia Perrelli ◽  
...  

Obestatin, a newly discovered peptide encoded by the ghrelin gene, induces the expression of genes regulating pancreatic β-cell differentiation, insulin biosynthesis, and glucose metabolism. It also activates antiapoptotic signaling pathways such as phosphoinositide 3-kinase (PI3K) and ERK1/2 in pancreatic β-cells and human islets. Since these kinases have been shown to protect against myocardial injury, we sought to investigate whether obestatin would exert cardioprotective effects. Both isolated perfused rat heart and cultured cardiomyocyte models of ischemia-reperfusion (I/R) were used to measure infarct size and cell apoptosis as end points of injury. The presence of specific obestatin receptors on cardiac cells as well as the signaling pathways underlying the obestatin effect were also studied. In the isolated heart, the addition of rat obestatin-(1–23) before ischemia reduced infarct size and contractile dysfunction in a concentration-dependent manner, whereas obestatin-(23–1), a synthetic analog with an inverse aminoacid sequence, was ineffective. The cardioprotective effect of obestatin-(1–23) was observed at concentrations of 10–50 nmol/l and was abolished by inhibiting PI3K or PKC by the addition of wortmannin (100 nmol/l) or chelerythrine, (5 μmol/l), respectively. In rat H9c2 cardiac cells or isolated ventricular myocytes subjected to I/R, 50 nmol/l obestatin-(1–23) reduced cardiomyocyte apoptosis and reduced caspase-3 activation; the antiapoptotic effect was blocked by the inhibition of PKC, PI3K, or ERK1/2 pathways. In keeping with these functional findings, radioreceptor binding results revealed the presence of specific high-affinity obestatin-binding sites, mainly localized on membranes of the ventricular myocardium and cardiomyocytes. Our data suggest that, by acting on specific receptors, obestatin-(1–23) activates PI3K, PKC-ε, PKC-δ, and ERK1/2 signaling and protects cardiac cells against myocardial injury and apoptosis induced by I/R.

2009 ◽  
Vol 297 (6) ◽  
pp. H2035-H2043 ◽  
Author(s):  
Sophie Tamareille ◽  
Nehmat Ghaboura ◽  
Frederic Treguer ◽  
Dalia Khachman ◽  
Anne Croué ◽  
...  

Ischemic postconditioning (IPost) and erythropoietin (EPO) have been shown to attenuate myocardial reperfusion injury using similar signaling pathways. The aim of this study was to examine whether EPO is as effective as IPost in decreasing postischemic myocardial injury in both Langendorff-isolated-heart and in vivo ischemia-reperfusion rat models. Rat hearts were subjected to 25 min ischemia, followed by 30 min or 2 h of reperfusion in the isolated-heart study. Rats underwent 45 min ischemia, followed by 24 h of reperfusion in the in vivo study. In both studies, the control group ( n = 12; ischemia-reperfusion only) was compared with IPost ( n = 16; 3 cycles of 10 s reperfusion/10 s ischemia) and EPO ( n = 12; 1,000 IU/kg) at the onset of reperfusion. The following resulted. First, in the isolated hearts, IPost or EPO significantly improved postischemic recovery of left ventricular developed pressure. EPO induced better left ventricular developed pressure than IPost at 30 min of reperfusion (73.18 ± 10.23 vs. 48.11 ± 7.92 mmHg, P < 0.05). After 2 h of reperfusion, the infarct size was significantly lower in EPO-treated hearts compared with IPost and control hearts (14.36 ± 0.60%, 19.11 ± 0.84%, and 36.21 ± 4.20% of the left ventricle, respectively; P < 0.05). GSK-3β phosphorylation, at 30 min of reperfusion, was significantly higher with EPO compared with IPost hearts. Phosphatidylinositol 3-kinase and ERK1/2 inhibitors abolished both EPO- and IPost-mediated cardioprotection. Second, in vivo, IPost and EPO induced an infarct size reduction compared with control (40.5 ± 3.6% and 28.9 ± 3.1%, respectively, vs. 53.7 ± 4.3% of the area at risk; P < 0.05). Again, EPO decreased significantly more infarct size and transmurality than IPost ( P < 0.05). In conclusion, with the use of our protocols, EPO showed better protective effects than IPost against reperfusion injury through higher phosphorylation of GSK-3β.


Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Carol Chen-Scarabelli ◽  
Zhaokan Yuan ◽  
Giuseppe Faggian ◽  
Francesco Santini ◽  
Alessio Rungatscher ◽  
...  

BACKGROUND: Cardioplegic arrest and subsequent reperfusion inevitably expose the heart to an iatrogenic ischemia/reperfusion injury (iIRI). We previously reported that iIRI caused mitochondria-initiated myocyte apoptosis, but also induction of urocortin (Ucn), an endogenous cardioprotective peptide. We also showed that Ucn induced PKCϵ-mediated opening of mitochondrial K ATP channels in isolated heart mitochondria. AIM: To investigate, in patients exposed to iIRI, the cardioprotective role and the mechanism of action of Ucn, with respect to PKCϵ expression, activation and relocation. METHODS AND RESULTS: Two sequential biopsies were obtained from the right atrium of 25 patients undergoing coronary artery bypass grafting at the start of grafting (internal control) and 10 mins after release of the aortic clamp. Mean values of ejection fraction, aortic cross-clamping time and number of grafts were 51±8; 48±8 mins; and 3.6±0.5 respectively. In hearts exposed to iIRI, RT-PCR and immunostaining showed Ucn induction at the mRNA (255% of basic levels, p<0.05) and protein level (28±2.1% positive myocytes vs 3.1±0.6% of internal control; p<0.01) respectively. iIRI also induced a selective increase of PKC-ϵ mRNA (225% of internal control; p<0.05) and a two-fold overexpression of total PKCϵ isoform (assessed by Western blotting; p<0.05), which paralleled a 2.9 fold increase in PKCϵ phosphorylation (p<0.01). TUNEL positivity (<0.1% and 2.9±0.7% positive myocytes pre- and post-iIRI respectively; p<0.01) was only seen in Ucn-negative cells, and, of note, Ucn-positive myocytes showed concurrent mitochondrial relocation of phosphorylated PKCϵ, as documented by mitochondrial-activated PKCϵ colocalization, calculated by confocal microscopy with an image analyzer software (% overlap: 57±5 vs 11±2 in Ucn-negative cells; p<0.01). Western blotting carried out in pools of cytosolic and mitochondrial fractions confirmed a 2.5 fold increase in mitochondrial localization of phosphorylated PKC-ϵ following iIRI (p<0.05). CONCLUSIONS : In patients exposed to iIRI, Ucn expression in viable cells was selectively associated with phosphorylation and mitochondrial relocation of PKCϵ, suggesting a cardioprotective role for endogenous Ucn in the human heart.


2013 ◽  
Vol 113 (suppl_1) ◽  
Author(s):  
Eric R Gross ◽  
Travis J Urban ◽  
Ana K Hsu ◽  
Nir Qvit ◽  
Garrett J Gross ◽  
...  

Introduction: The transient receptor potential 1 channel (TRPV1) mediates signals from pain, heat, and/or noxious stimuli. TRPV1 sensitization can occur via a protein kinase C (PKC)-dependent mechanism in neurons. Therefore, we tested whether TRPV1 is a mediator of cardioprotection in models of ischemia-reperfusion and whether the molecular mechanism of cardioprotection occurs via PKC-induced TRPV1 channel sensitization. Methods: Male Sprague Dawley rats and H9C2 left ventricle-derived cells were used for whole animal and cellular ischemia-reperfusion studies to test this hypothesis. Statistical analysis regarding infarct size, calculated as percentage of area at risk per left ventricle, was performed by a one way ANOVA (*P<0.01). Results: Remote preconditioning-induced infarct size reduction via an abdominal surgical incision was blocked by prior administration of a selective TRPV1 peptide inhibitor, V1-B (3.0mg/kg), given over the incision site (Incision: 44±2*% V1-B+Incision: 65±2% versus Control: 64±1% n=6/group). Capsaicin (0.3mg/kg) given intravenously through the internal jugular vein reduced infarct size in vivo , which was blocked by prior capsazepine (TRPV1 inhibitor, 3.0mg/kg) administration (Capsaicin: 43±2* Capsaicin+ capsazepine: 64±4 versus Control: 62±3, n=7/group). Further in an ex vivo isolated heart model, infarct size reduction afforded by the selective epsilon PKC activator (pseudo epsilon RACK, 1uM) was partially blocked with prior treatment of V1-B (1uM), the TRPV1 peptide blocker (pseudo epsilon RACK: 20±2*%, pseudo epsilon RACK+V1-B: 42±4% versus control: 47±4%, n=7/group). TRPV1 expression was found in both whole heart homogenate and in the H9C2 cell line. Using a model of ischemia-reoxygenation in H9C2 cells, capsaicin treatment before and during ischemia-reoxygenation reduced cellular damage as assessed by MTT and LDH assays. Greater damage occurred with TRPV1 inhibition by capsazepine compared to control. Conclusions: Our studies suggest TRPV1 contributes an essential role for both remote and direct cardioprotection. Further studies are ongoing to determine the post-translational sites on TRPV1 and how a TRPV1-epsilon PKC protein-protein interaction induces cardioprotection.


2019 ◽  
Vol 97 (12) ◽  
pp. 1124-1131 ◽  
Author(s):  
Aysu Kilic ◽  
Savas Ustunova ◽  
Cansu Usta ◽  
Huri Bulut ◽  
Ismail Meral ◽  
...  

Our study aimed to determine the effects of losartan and PD123319 in ischemia–reperfusion (IR) injury in isolated perfused rat heart. The study used 40 male Wistar albino rats that were grouped as Control, IR, and IR treatment groups that received losartan (20 mg/kg), PD123319 (20 mg/kg), and losartan+PD123319. The hearts were attached to Langendorff isolated heart system by employing in situ cannulation method, and cardiodynamic parameters were recorded during the experiment. At the end of experiment, hearts were retained for biochemical analysis and all data were statistically evaluated. A partial recovery of cardiodynamic parameters was observed in all treatment groups. A significant increase in oxidative stress parameters were seen in the IR group, whereas all treatment groups exhibited lower increase. Furthermore, levels of all antioxidant parameters were significantly lower in the IR group, but higher in the treatment groups. Effects on all parameters were much more remarkable in the PD123319 group. Levels of angiotensin II and renin were increased (P < 0.001) with IR application and decreased (P < 0.001) with the treatment of both antagonists. In conclusion, treatment of losartan and PD123319 played a cardioprotective role against IR injury, PD123319 being more effective in this protection.


2008 ◽  
Vol 294 (6) ◽  
pp. H2473-H2479 ◽  
Author(s):  
Seiji Matsuhisa ◽  
Hajime Otani ◽  
Toru Okazaki ◽  
Koji Yamashita ◽  
Yuzo Akita ◽  
...  

Oxidative stress is involved in the tolerance to ischemia-reperfusion (I/R) injury. Because angiotensin II type 1 receptor blockers (ARBs) inhibit oxidative stress, there is concern that ARBs abolish the tolerance to I/R injury. Dahl salt-sensitive (DS) hypertensive and salt-resistant (DR) normotensive rats received an antioxidant, 2-mercaptopropionylglycine (MPG), or an ARB, losartan, for 7 days. Losartan and MPG significantly inhibited oxidative stress as determined by tissue malondialdehyde + 4-hydroxynoneal and increased expression of inducible nitric oxide synthase (iNOS) in the DS rat heart. However, losartan but not MPG activated endothelial nitric oxide synthase (eNOS) as assessed by phosphorylation of eNOS on Ser1177. Infarct size after 30-min left coronary artery occlusion followed by 2-h reperfusion was comparable between DS and DR rat hearts. Although MPG and losartan had no effect on infarct size in the DR rat heart, MPG but not losartan significantly increased infarct size in the DS rat heart. A selective iNOS inhibitor, 1400W, increased infarct size in the DS rat heart, but it had no effect on infarct size in the losartan-treated DS rat heart. However, a nonselective NOS inhibitor, Nω-nitro-l-arginine methyl ester, increased infarct size in the losartan-treated DS rat heart. These results suggest that losartan preserves the tolerance to I/R injury by activating eNOS despite elimination of redox-sensitive upregulation of iNOS and iNOS-dependent cardioprotection in the DS rat heart.


2018 ◽  
Vol 23 (5) ◽  
pp. 446-455 ◽  
Author(s):  
Oussama Bakhta ◽  
Simon Blanchard ◽  
Anne-Laure Guihot ◽  
Sophie Tamareille ◽  
Delphine Mirebeau-Prunier ◽  
...  

Background: Inflammation plays a crucial role in the pathophysiology of myocardial ischemia/reperfusion (I/R) injury. A clinical trial has recently reported a smaller infarct size in a cohort of patients with ST-segment elevation myocardial infarction (MI) treated with a short colchicine course. The mechanism underlying colchicine-induced cardioprotection in the early MI phase remains unclear. We hypothesized that a short pretreatment with colchicine could induce acute beneficial effects by protecting the heart against inflammation in myocardial I/R injury. Methods and Results: Rats were subjected to 40-minute left anterior descending coronary occlusion, followed by 120-minute reperfusion. Colchicine (0.3 mg/kg) or a vehicle was administered per os 24 hours and immediately before surgery. Infarct size was significantly reduced in the colchicine group (35.6% ± 3.0% vs 46.6% ± 3.3%, P < .05). The beneficial effects of colchicine were associated with an increased systemic interleukin-10 (IL-10) level and decreased cardiac transforming growth factor-β level. Interleukin-1β was found to increase in a “time of reperfusion”-dependent manner. Colchicine inhibited messenger RNA expression of caspase-1 and pro-IL-18. Interleukin-1β injected 10 minutes prior to myocardial ischemia induced greater infarct size (58.0% ± 2.0%, P < .05) as compared to the vehicle. Colchicine combined to IL-1β injection significantly decreased infarct size (47.1% ± 2.2%, P < .05) as compared to IL-1β alone, while colchicine alone exhibited a significantly more marked cardioprotective effect than the colchicine-IL-1β association. Conclusion: The cardioprotection induced by a short colchicine pretreatment was associated with an anti-inflammatory effect in the early reperfusion phase in our rat MI model.


2003 ◽  
Vol 284 (5) ◽  
pp. H1612-H1617 ◽  
Author(s):  
Hongjiang Chen ◽  
Dayuan Li ◽  
Tom Saldeen ◽  
Jawahar L. Mehta

Ischemia-reperfusion (I/R) is thought to upregulate the expression and activity of matrix metalloproteinases (MMPs), which regulate myocardial and vascular remodeling. Previous studies have shown that transforming growth factor-β1 (TGF-β1) can attenuate myocardial injury induced by I/R. TGF-β1 is also reported to suppress the release of MMPs. To study the modulation of MMP-1 by TGF-β1 in I/R myocardium, Sprague-Dawley rats were given saline and subjected to 1 h of myocardial ischemia [total left coronary artery (LCA) ligation] followed by 1 h of reperfusion ( n = 9). Parallel groups of rats were pretreated with recombinant TGF-β1(rTGF-β1, 1 mg/rat, n = 9) before reperfusion or exposure to sham I/R (control group). I/R caused myocardial necrosis and dysfunction, indicated by decreased first derivative of left ventricular pressure, mean arterial blood pressure, and heart rate (all P < 0.01 vs. sham-operated control group). Simultaneously, I/R upregulated MMP-1 ( P < 0.01). Treatment of rats with rTGF-β1 reduced the extent of myocardial necrosis and dysfunction despite I/R (all P < 0.01). rTGF-β1 treatment also inhibited the upregulation of MMP-1 in the I/R myocardium ( P < 0.05). To determine the direct effect of MMP-1 on the myocardium, isolated adult rat myocytes were treated with active MMP-1, which caused injury and death of cultured myocytes, measured as lactate dehydrogenase release and trypan blue staining, in a dose- and time-dependent manner ( P < 0.05). Pretreatment with PD-166793, a specific MMP inhibitor, attenuated myocardial injury and death induced by active MMP-1. The present study for the first time shows that MMP-1 can directly cause myocyte injury or death and that attenuation of myocardial I/R injury by TGF-β1 may, at least partly, be mediated by the inhibition of upregulation of MMP-1.


Antioxidants ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 1667
Author(s):  
Yan-Cheng Shen ◽  
Yan-Jhih Shen ◽  
Wen-Sen Lee ◽  
Michael Yu-Chih Chen ◽  
Wei-Chia Tu ◽  
...  

To identify the core structure of 2-aminoethoxydiphenyl borate (2-APB) responsible for the anti-oxidative and protective effect on the ischemia/reperfusion (I/R)-induced heart injury, various 2-APB analogues were analyzed, and several antioxidant assays were performed. Cell viability was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Myocardial infarct size was quantified using triphenyl tetrazolium chloride (TTC) staining. The levels of tumor necrosis factor-alpha (TNF-α) and cleaved-caspase-3 protein were evaluated as an indicator for the anti-inflammatory and anti-apoptotic effect, respectively. Our data show that 2-APB, diphenylborinic anhydride (DPBA) and 3-(diphenylphosphino)-1-propylamine (DP3A) all exerted the anti-oxidative activity, but only 2-APB and DPBA can scavenge H2O2. 2-APB and DPBA can potently inhibit hydrogen peroxide (H2O2)- and hypoxanthine/xanthine oxidase (HX/XOD)-induced increases in intracellular H2O2 and H9c2 cell death. 2-APB and DPBA were able to decrease the I/R-induced adult rat cardiomyocytes death, myocardial infarct size, and the levels of malondialdehyde (MDA) and creatine kinase-MB (CK-MB). Our results suggest that the two benzene rings with a boron atom comprise the core structure of 2-APB responsible for the anti-oxidative effect mediated through the reaction with H2O2 and generation of phenolic compounds, which in turn reduced the I/R-induced oxidative stress and injury in the rat heart.


Author(s):  
Gang Zhang ◽  
Kun Yu ◽  
Zhi Bao ◽  
Xiaofeng Sun ◽  
Dongying Zhang

Background. Ischemia/reperfusion (I/R) induced lethal tissue injury in myocardium. FoxM1 (Forkhead Box M1), expressed in proliferating cardiac progenitor cells, could regulate myocardial development. However, the role of FoxM1 in I/R-induced myocardial injury has not been reported yet. Methods. Rats were conducted with regional ischemia followed by reperfusion in myocardium through ligation of the left anterior descending coronary artery. Triphenyl-tetrazolium chloride staining was utilized to assess the infarct size. ELISA was performed to detect activities of creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH). Protein expression of FoxM1 in heart tissues and H9c2 were determined by western blot. H9c2 cells were used to establish a hypoxia/reoxygenation cell model, and the cell viability, proliferation and apoptosis were evaluated by MTT, EdU (5-ethynyl-2’-deoxyuridine) staining and TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, respectively. Adenovirus (Ad)-mediated over-expression of FoxM1 was injected into the anterior wall of the left ventricle of rats to evaluate the role of FoxM1 on in vivo I/R-induced myocardial injury. Results. FoxM1 was reduced in heart tissues isolated from rats post myocardial I/R injury. Forced FoxM1 expression increased cell viability and proliferation of hypoxia/reoxygenation-induced H9c2, while repressed the cell apoptosis with increased Bcl-2 and decreased Bax and cleaved caspase-3. Injection of Ad-FoxM1 suppressed infarct size of the heart and decreased activities of CK-MB and LDH. Conclusion. FoxM1 attenuated I/R-induced myocardial injury, providing potential therapeutic target for the disease.


Sign in / Sign up

Export Citation Format

Share Document