301
|
Yu M, Wen N, Wenzhong Z, Yuanchang X, Xiaoming D, Yongjin L. Effect of repeated ischaemic preconditioning on TLR4 and proinflammatory cytokines TNF-α and IL-1β in myocardial ischaemia-reperfusion injury in a rat model. Arch Med Sci 2010; 6:843-7. [PMID: 22427755 PMCID: PMC3302693 DOI: 10.5114/aoms.2010.19289] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 09/12/2010] [Accepted: 10/05/2010] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION The role of TLR4 in ischaemic preconditioning is still unclear; we do not know the change of the expression of TLR4 in the process. In this study, we used ischaemic preconditioning models to observe the change of TLR4 expression and the level of proinflammatory cytokines TNF-α and IL-1β to investigate the protective mechanism of TLR4 in ischaemic preconditioning for myocardial ischaemia-reperfusion injury (MI/RI) in rats. MATERIAL AND METHODS Eighteen male Sprague-Dawley (SD) rats were randomly separated into sham, ischaemic reperfusion (IR) and ischaemic preconditioning (IP) groups (6/group). Peripheral blood and cardiac muscle with pathological changes were collected after the establishment of the above three animal models. We used ELISA to determine proinflammatory cytokines TNF-α and IL-1β production in serum of these animals. RT-PCR and Western blot were used to assay the transcriptional level and protein level of TLR4 in cardiac muscle tissue with pathological changes, respectively. RESULTS We found that compared with the IR group, ischaemic preconditioning could effectively reduce the expression levels of TNF-α and IL-1β in sera of rats in the IP group (p < 0.01). Meanwhile, TLR4 mRNA and protein levels were down-regulated (p < 0.01 and p < 0.05, respectively). We also found that infarct size decreased in the IP group compared with the IR group (p < 0.05). CONCLUSIONS Based on the results, we can conclude that the specific mechanism of ischaemic preconditioning for RI might be closely associated with decreasing expression levels of TLR4 and proinflammatory cytokines such as TNF-α and IL-1β.
Collapse
Affiliation(s)
- Ma Yu
- Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ni Wen
- Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhu Wenzhong
- Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiong Yuanchang
- Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Deng Xiaoming
- Department of Anesthesia, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Luo Yongjin
- Department of Cerebral Surgery, Changhai Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
302
|
Catestatin improves post-ischemic left ventricular function and decreases ischemia/reperfusion injury in heart. Cell Mol Neurobiol 2010; 30:1171-9. [PMID: 21104119 PMCID: PMC3008938 DOI: 10.1007/s10571-010-9598-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 09/02/2010] [Indexed: 02/02/2023]
Abstract
The Chromogranin A (CgA)-derived anti-hypertensive peptide catestatin (CST) antagonizes catecholamine secretion, and is a negative myocardial inotrope acting via a nitric oxide-dependent mechanism. It is not known whether CST contributes to ischemia/reperfusion injury or is a component of a cardioprotective response to limit injury. Here, we tested whether CST by virtue of its negative inotropic activity improves post-ischemic cardiac function and cardiomyocyte survival. Three groups of isolated perfused hearts from adult Wistar rats underwent 30-min ischemia and 120-min reperfusion (I/R, Group 1), or were post-conditioned by brief ischemic episodes (PostC, 5-cycles of 10-s I/R at the beginning of 120-min reperfusion, Group 2), or with exogenous CST (75 nM for 20 min, CST-Post, Group-3) at the onset of reperfusion. Perfusion pressure and left ventricular pressure (LVP) were monitored. Infarct size was evaluated with nitroblue-tetrazolium staining. The CST (5 nM) effects were also tested in simulated ischemia/reperfusion experiments on cardiomyocytes isolated from young-adult rats, evaluating cell survival with propidium iodide labeling. Infarct size was 61 ± 6% of risk area in hearts subjected to I/R only. PostC reduced infarct size to 34 ± 5%. Infarct size in CST-Post was 36 ± 3% of risk area (P < 0.05 respect to I/R). CST-Post reduced post-ischemic rise of diastolic LVP, an index of contracture, and significantly improved post-ischemic recovery of developed LVP. In isolated cardiomyocytes, CST increased the cell viability rate by about 65% after simulated ischemia/reperfusion. These results suggest a novel cardioprotective role for CST, which appears mainly due to a direct reduction of post-ischemic myocardial damages and dysfunction, rather than to an involvement of adrenergic terminals and/or endothelium.
Collapse
|
303
|
Remote postconditioning by humoral factors in effluent from ischemic preconditioned rat hearts is mediated via PI3K/Akt-dependent cell-survival signaling at reperfusion. Basic Res Cardiol 2010; 106:135-45. [PMID: 21103992 PMCID: PMC3012213 DOI: 10.1007/s00395-010-0133-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 10/19/2010] [Accepted: 11/02/2010] [Indexed: 12/12/2022]
Abstract
Short non-lethal ischemic episodes administered to hearts prior to (ischemic preconditioning, IPC) or directly after (ischemic postconditioning, IPost) ischemic events facilitate myocardial protection. Transferring coronary effluent collected during IPC treatment to un-preconditioned recipient hearts protects from lethal ischemic insults. We propose that coronary IPC effluent contains hydrophobic cytoprotective mediators acting via PI3K/Akt-dependent pro-survival signaling at ischemic reperfusion. Ex vivo rat hearts were subjected to 30 min of regional ischemia and 120 min of reperfusion. IPC effluent administered for 10 min prior to index ischemia attenuated infarct size by ≥55% versus control hearts (P < 0.05). Effluent administration for 10 min at immediate reperfusion (reperfusion therapy) or as a mimetic of pharmacological postconditioning (remote postconditioning, RIPost) significantly reduced infarct size compared to control (P < 0.05). The IPC effluent significantly increased Akt phosphorylation in un-preconditioned hearts when administered before ischemia or at reperfusion, while pharmacological inhibition of PI3K/Akt-signaling at reperfusion completely abrogated the cardioprotection offered by effluent administration. Fractionation of coronary IPC effluent revealed that cytoprotective humoral mediator(s) released during the conditioning phase were of hydrophobic nature as all hydrophobic fractions with molecules under 30 kDa significantly reduced infarct size versus the control and hydrophilic fraction-treated hearts (P < 0.05). The total hydrophobic effluent fraction significantly reduced infarct size independently of temporal administration (before ischemia, at reperfusion or as remote postconditioning). In conclusion, the IPC effluent retains strong cardioprotective properties, containing hydrophobic mediator(s) < 30 kDa offering cytoprotection via PI3K/Akt-dependent signaling at ischemic reperfusion.
Collapse
|
304
|
Lethal myocardial reperfusion injury: a necessary evil? Int J Cardiol 2010; 151:3-11. [PMID: 21093938 DOI: 10.1016/j.ijcard.2010.10.056] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 10/23/2010] [Accepted: 10/27/2010] [Indexed: 12/11/2022]
Abstract
Despite being the most effective means of limiting infarct size, coronary reperfusion comes at a price and induces additional damage to the myocardium. Lethal reperfusion injury (death of myocytes that were viable at the time of reperfusion) is an increasingly acknowledged phenomenon. There are many interconnected mechanisms involved in this type of cell death. Calcium overload (generating myocyte hypercontracture), rapid recovery of physiological pH, neutrophil infiltration of the ischemic area, opening of the mitochondrial permeability-transition-pore (PTP), and apoptotic cell death are among the more important mechanisms involved in reperfusion injury. The activation of a group of proteins called reperfusion injury salvage kinases (RISK) pathway confers protection against reperfusion injury, mainly by inhibiting the opening of the mitochondrial PTP. Many interventions have been tested in human trials triggered by encouraging animal studies. In the present review we will explain in detail the main mechanism involved in reperfusion injury, as well as the various approaches (pre-clinical and human trials) performed targeting these mechanisms. Currently, no intervention has been consistently shown to reduce reperfusion injury in large randomized multicenter trials, but the research in this field is intense and the future is highly promising.
Collapse
|
305
|
Jaffe R, Dick A, Strauss BH. Prevention and treatment of microvascular obstruction-related myocardial injury and coronary no-reflow following percutaneous coronary intervention: a systematic approach. JACC Cardiovasc Interv 2010; 3:695-704. [PMID: 20650430 DOI: 10.1016/j.jcin.2010.05.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 05/10/2010] [Accepted: 05/12/2010] [Indexed: 11/18/2022]
Abstract
Microvascular obstruction (MVO) commonly occurs following percutaneous coronary interventions (PCI), may lead to myocardial injury, and is an independent predictor of adverse outcome. Severe MVO may manifest angiographically as reduced flow in the patent upstream epicardial arteries, a situation that is termed "no-reflow." Microvascular obstruction can be broadly categorized according to the duration of myocardial ischemia preceding PCI. In "interventional MVO" (e.g., elective PCI), obstruction typically involves myocardium that was not exposed to acute ischemia before PCI. Conversely "reperfusion MVO" (e.g., primary PCI for acute myocardial infarction) occurs within a myocardial territory that was ischemic before the coronary intervention. Interventional and reperfusion MVO have distinct pathophysiological mechanisms and may require individualized therapeutic approaches. Interventional MVO is triggered predominantly by downstream embolization of atherosclerotic material from the epicardial vessel wall into the distal microvasculature. Reperfusion MVO results from both distal embolization and ischemia-reperfusion injury within the subtended ischemic tissue. Management of MVO and no-reflow may be targeted at different levels: the epicardial artery, microvasculature, and tissue. The aim of the present report is to advocate a systematic approach to prevention and treatment of MVO in different clinical settings. Randomized clinical trials have studied strategies for prevention of MVO and no-reflow; however, the efficacy of measures for reversing MVO once no-reflow has been demonstrated angiographically is unclear. New approaches for prevention and treatment of MVO will require a better understanding of intracellular cardioprotective pathways such as the blockade of the mitochondrial permeability transition pore.
Collapse
Affiliation(s)
- Ronen Jaffe
- Lady Davis Medical Center, Department of Cardiology, Haifa, Israel.
| | | | | |
Collapse
|
306
|
Boengler K, Hilfiker-Kleiner D, Heusch G, Schulz R. Inhibition of permeability transition pore opening by mitochondrial STAT3 and its role in myocardial ischemia/reperfusion. Basic Res Cardiol 2010; 105:771-85. [PMID: 20960209 PMCID: PMC2978889 DOI: 10.1007/s00395-010-0124-1] [Citation(s) in RCA: 312] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 09/27/2010] [Accepted: 10/01/2010] [Indexed: 01/17/2023]
Abstract
The signal transducer and activator of transcription 3 (STAT3) contributes to cardioprotection by ischemic pre- and postconditioning. Mitochondria are central elements of cardioprotective signaling, most likely by delaying mitochondrial permeability transition pore (MPTP) opening, and STAT3 has recently been identified in mitochondria. We now characterized the mitochondrial localization of STAT3 and its impact on respiration and MPTP opening. STAT3 was mainly present in the matrix of subsarcolemmal and interfibrillar cardiomyocyte mitochondria. STAT1, but not STAT5 was also detected in mitochondria under physiological conditions. ADP-stimulated respiration was reduced in mitochondria from mice with a cardiomyocyte-specific deletion of STAT3 (STAT3-KO) versus wildtypes and in rat mitochondria treated with the STAT3 inhibitor Stattic (STAT3 inhibitory compound, 6-Nitrobenzo[b]thiophene 1,1-dioxide). Mitochondria from STAT3-KO mice and Stattic-treated rat mitochondria tolerated less calcium until MPTP opening occurred. STAT3 co-immunoprecipitated with cyclophilin D, the target of the cardioprotective agent and MPTP inhibitor cyclosporine A (CsA). However, CsA reduced infarct size to a similar extent in wildtype and STAT3-KO mice in vivo. Thus, STAT3 possibly contributes to cardioprotection by stimulation of respiration and inhibition of MPTP opening.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institut für Pathophysiologie, Zentrum für Innere Medizin, Universitätsklinikum Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Denise Hilfiker-Kleiner
- Molekulare Kardiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Gerd Heusch
- Institut für Pathophysiologie, Zentrum für Innere Medizin, Universitätsklinikum Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Rainer Schulz
- Institut für Pathophysiologie, Zentrum für Innere Medizin, Universitätsklinikum Essen, Hufelandstr. 55, 45122 Essen, Germany
| |
Collapse
|
307
|
Ghaboura N, Tamareille S, Ducluzeau PH, Grimaud L, Loufrani L, Croué A, Tourmen Y, Henrion D, Furber A, Prunier F. Diabetes mellitus abrogates erythropoietin-induced cardioprotection against ischemic-reperfusion injury by alteration of the RISK/GSK-3β signaling. Basic Res Cardiol 2010; 106:147-62. [PMID: 20981553 DOI: 10.1007/s00395-010-0130-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Revised: 10/18/2010] [Accepted: 10/20/2010] [Indexed: 12/22/2022]
Abstract
Recent studies reported cardioprotective effects of erythropoietin (EPO) against ischemia-reperfusion (I/R) injury through activation of the reperfusion injury salvage kinase (RISK) pathway. As RISK has been reported to be impaired in diabetes and insulin resistance syndrome, we examined whether EPO-induced cardioprotection was maintained in rat models of type 1 diabetes and insulin resistance syndrome. Isolated hearts were obtained from three rat cohorts: healthy controls, streptozotocin (STZ)-induced diabetes, and high-fat diet (HFD)-induced insulin resistance syndrome. All hearts underwent 25 min ischemia and 30 min or 120 min reperfusion. They were assigned to receive either no intervention or a single dose of EPO at the onset of reperfusion. In hearts from healthy controls, EPO decreased infarct size (14.36 ± 0.60 and 36.22 ± 4.20% of left ventricle in EPO-treated and untreated hearts, respectively, p < 0.05) and increased phosphorylated forms of Akt, ERK1/2, and their downstream target GSK-3β. In hearts from STZ-induced diabetic rats, EPO did not decrease infarct size (32.05 ± 2.38 and 31.88 ± 1.87% in EPO-treated and untreated diabetic rat hearts, respectively, NS) nor did it increase phosphorylation of Akt, ERK1/2, and GSK-3β. In contrast, in hearts from HFD-induced insulin resistance rats, EPO decreased infarct size (18.66 ± 1.99 and 34.62 ± 3.41% in EPO-treated and untreated HFD rat hearts, respectively, p < 0.05) and increased phosphorylation of Akt, ERK1/2, and GSK-3β. Administration of GSK-3β inhibitor SB216763 was cardioprotective in healthy and diabetic hearts. STZ-induced diabetes abolished EPO-induced cardioprotection against I/R injury through a disruption of upstream signaling of GSK-3β. In conclusion, direct inhibition of GSK-3β may provide an alternative strategy to protect diabetic hearts against I/R injury.
Collapse
Affiliation(s)
- Nehmat Ghaboura
- Protection et Remodelage du Myocarde, UPRES EA 3860, Faculté de Médecine, Université d'Angers, Rue Haute de Reculée, 49045 Angers Cedex 1, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
308
|
Gao E, Lei YH, Shang X, Huang ZM, Zuo L, Boucher M, Fan Q, Chuprun JK, Ma XL, Koch WJ. A novel and efficient model of coronary artery ligation and myocardial infarction in the mouse. Circ Res 2010; 107:1445-53. [PMID: 20966393 DOI: 10.1161/circresaha.110.223925] [Citation(s) in RCA: 522] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RATIONALE coronary artery ligation to induce myocardial infarction (MI) in mice is typically performed by an invasive and time-consuming approach that requires ventilation and chest opening (classic method), often resulting in extensive tissue damage and high mortality. We developed a novel and rapid surgical method to induce MI that does not require ventilation. OBJECTIVE the purpose of this study was to develop and comprehensively describe this method and directly compare it to the classic method. METHODS AND RESULTS male C57/B6 mice were grouped into 4 groups: new method MI (MI-N) or sham (S-N) and classic method MI (MI-C) or sham (S-C). In the new method, heart was manually exposed without intubation through a small incision and MI was induced. In the classic method, MI was induced through a ventilated thoracotomy. Similar groups were used in an ischemia/reperfusion injury model. This novel MI procedure is rapid, with an average procedure time of 1.22 ± 0.05 minutes, whereas the classic method requires 23.2 ± 0.6 minutes per procedure. Surgical mortality was 3% in MI-N and 15.9% in MI-C. The rate of arrhythmia was significantly lower in MI-N. The postsurgical levels of tumor necrosis factor-α and myeloperoxidase were lower in new method, indicating less inflammation. Overall, 28-day post-MI survival rate was 68% with MI-N and 48% with MI-C. Importantly, there was no difference in infarct size or post-MI cardiac function between the methods. CONCLUSIONS this new rapid method of MI in mice represents a more efficient and less damaging model of myocardial ischemic injury compared with the classic method.
Collapse
Affiliation(s)
- Erhe Gao
- Center for Translational Medicine, George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Thomas Jefferson University, 1025 Walnut St, Room 302, Philadelphia, PA 19107, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
309
|
Kelly RF, Lamont KT, Somers S, Hacking D, Lacerda L, Thomas P, Opie LH, Lecour S. Ethanolamine is a novel STAT-3 dependent cardioprotective agent. Basic Res Cardiol 2010; 105:763-70. [PMID: 20938668 DOI: 10.1007/s00395-010-0125-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 10/01/2010] [Accepted: 10/05/2010] [Indexed: 01/09/2023]
Abstract
Ethanolamine is a biogenic amine found naturally in the body as part of membrane lipids and as a metabolite of the cardioprotective substances, sphingosine-1-phosphate (S1P) and anandamide. In the brain, ethanolamine, formed from the breakdown of anandamide protects against ischaemic apoptosis. However, the effects of ethanolamine in the heart are unknown. Signal transducer and activator of transcription 3 (STAT-3) is a critical prosurvival factor in ischaemia/reperfusion (I/R) injury. Therefore, we investigated whether ethanolamine protects the heart via activation of STAT-3. Isolated hearts from wildtype or cardiomyocyte specific STAT-3 knockout (K/O) mice were pre-treated with ethanolamine (Etn) (0.3 mmol/L) before I/R insult. In vivo rat hearts were subjected to 30 min ischaemia/2 h reperfusion in the presence or absence of 5 mg/kg S1P and/or the FAAH inhibitor, URB597. Infarct size was measured at the end of each protocol by triphenyltetrazolium chloride staining. Pre-treatment with ethanolamine decreased infarct size in isolated mouse or rat hearts subjected to I/R but this infarct sparing effect was lost in cardiomyocyte specific STAT-3 deficient mice. Pre-treatment with ethanolamine increased nuclear phosphorylated STAT-3 [control 0.75 ± 0.08 vs. Etn 1.50 ± 0.09 arbitrary units; P < 0.05]. Our findings suggest a novel cardioprotective role for ethanolamine against I/R injury via activation of STAT-3.
Collapse
Affiliation(s)
- Roisin F Kelly
- Hatter Cardiovascular Research Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | | | | | | | | | | | | | | |
Collapse
|
310
|
Javadov S, Rajapurohitam V, Kilić A, Hunter JC, Zeidan A, Said Faruq N, Escobales N, Karmazyn M. Expression of mitochondrial fusion-fission proteins during post-infarction remodeling: the effect of NHE-1 inhibition. Basic Res Cardiol 2010; 106:99-109. [PMID: 20886221 DOI: 10.1007/s00395-010-0122-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 09/01/2010] [Accepted: 09/19/2010] [Indexed: 11/29/2022]
Abstract
Studies on the role of mitochondrial fission/fusion (MFF) proteins in the heart have been initiated recently due to their biological significance in cell metabolism. We hypothesized that the expression of MFF proteins is affected by post-infarction remodeling and in vitro cardiomyocyte hypertrophy, and serves as a target for the Na(+)/H(+) exchanger 1 (NHE-1) inhibition. Post-infarction remodeling was induced in Sprague-Dawley rats by coronary artery ligation (CAL) while in vitro hypertrophy was induced in cardiomyocytes by phenylephrine (PE). Mitochondrial fission (Fis1, DRP1) and fusion (Mfn2, OPA1) proteins were analyzed in heart homogenates and cell lysates by Western blotting. Our results showed that 12 and 18 weeks after CAL, Fis1 increased by 80% (P < 0.01) and 31% (P < 0.05), and Mfn2 was reduced by 17% (P < 0.05) and 22% (P < 0.05), respectively. OPA1 was not changed at 12 weeks, although its expression decreased by 18% (P < 0.05) with 18 weeks of ligation. MFF proteins were also affected by PE-induced hypertrophy that was dependent on mitochondrial permeability transition pore opening and oxidative stress. The NHE-1-specific inhibitor EMD-87580 (EMD) attenuated changes in the expression of MFF proteins in both the models of hypertrophy. The effect of EMD was likely mediated, at least in part, through its direct action on mitochondria since Percoll-purified mitochondria and mitoplasts have been shown to contain NHE-1. Our study provides the first evidence linking cardiac hypertrophy with MFF proteins expression that was affected by NHE-1 inhibition, thus suggesting that MFF proteins might be a target for pharmacotherapy with anti-hypertrophic drugs.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, USA.
| | | | | | | | | | | | | | | |
Collapse
|
311
|
Sala-Mercado JA, Wider J, Undyala VVR, Jahania S, Yoo W, Mentzer RM, Gottlieb RA, Przyklenk K. Profound cardioprotection with chloramphenicol succinate in the swine model of myocardial ischemia-reperfusion injury. Circulation 2010; 122:S179-84. [PMID: 20837911 DOI: 10.1161/circulationaha.109.928242] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Emerging evidence suggests that "adaptive" induction of autophagy (the cellular process responsible for the degradation and recycling of proteins and organelles) may confer a cardioprotective phenotype and represent a novel strategy to limit ischemia-reperfusion injury. Our aim was to test this paradigm in a clinically relevant, large animal model of acute myocardial infarction. METHODS AND RESULTS Anesthetized pigs underwent 45 minutes of coronary artery occlusion and 3 hours of reperfusion. In the first component of the study, pigs received chloramphenicol succinate (CAPS) (an agent that purportedly upregulates autophagy; 20 mg/kg) or saline at 10 minutes before ischemia. Infarct size was delineated by tetrazolium staining and expressed as a % of the at-risk myocardium. In separate animals, myocardial samples were harvested at baseline and 10 minutes following CAPS treatment and assayed (by immunoblotting) for 2 proteins involved in autophagosome formation: Beclin-1 and microtubule-associated protein light chain 3-II. To investigate whether the efficacy of CAPS was maintained with "delayed" treatment, additional pigs received CAPS (20 mg/kg) at 30 minutes after occlusion. Expression of Beclin-1 and microtubule-associated protein light chain 3-II, as well as infarct size, were assessed at end-reperfusion. CAPS was cardioprotective: infarct size was 25±5 and 41±4%, respectively, in the CAPS-pretreated and CAPS-delayed treatment groups versus 56±5% in saline controls (P<0.01 and P<0.05 versus control). Moreover, administration of CAPS was associated with increased expression of both proteins. CONCLUSIONS Our results demonstrate attenuation of ischemia-reperfusion injury with CAPS and are consistent with the concept that induction of autophagy may provide a novel strategy to confer cardioprotection.
Collapse
Affiliation(s)
- Javier A Sala-Mercado
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | |
Collapse
|
312
|
Translating novel strategies for cardioprotection: the Hatter Workshop Recommendations. Basic Res Cardiol 2010; 105:677-86. [PMID: 20865418 PMCID: PMC2965360 DOI: 10.1007/s00395-010-0121-4] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 09/09/2010] [Accepted: 09/10/2010] [Indexed: 12/18/2022]
Abstract
Ischemic heart disease (IHD) is the leading cause of death worldwide. Novel cardioprotective strategies are therefore required to improve clinical outcomes in patients with IHD. Although a large number of novel cardioprotective strategies have been discovered in the research laboratory, their translation to the clinical setting has been largely disappointing. The reason for this failure can be attributed to a number of factors including the inadequacy of the animal ischemia–reperfusion injury models used in the preclinical cardioprotection studies and the inappropriate design and execution of the clinical cardioprotection studies. This important issue was the main topic of discussion of the UCL-Hatter Cardiovascular Institute 6th International Cardioprotection Workshop, the outcome of which has been published in this article as the “Hatter Workshop Recommendations”. These have been proposed to provide guidance on the design and execution of both preclinical and clinical cardioprotection studies in order to facilitate the translation of future novel cardioprotective strategies for patient benefit.
Collapse
|
313
|
|
314
|
Zhuo C, Wang Y, Wang X, Wang Y, Chen Y. Cardioprotection by ischemic postconditioning is abolished in depressed rats: role of Akt and signal transducer and activator of transcription-3. Mol Cell Biochem 2010; 346:39-47. [PMID: 20830508 DOI: 10.1007/s11010-010-0589-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Accepted: 08/28/2010] [Indexed: 11/24/2022]
Abstract
Ischemic postconditioning (IPC) represents one of the most effective cardioprotective strategies against myocardial ischemia/reperfusion. Depression is commonly present in patients with coronary heart disease. However, whether depression interferes with the cardioprotection of IPC during myocardial ischemia/reperfusion and their underlying mechanisms remain largely unknown. Isolated hearts from chronic mild stress induced-depressed rats and non-depressed rats were subjected to 30 min of regional ischemia followed by 120 min of reperfusion in the presence or absence of IPC (consisting of 6 cycles of 10 s of reperfusion and 10 s of ischemia immediately after the sustained ischemia). Myocardial infarct size, creatine kinase (CK) and cardiac troponin T (cTnT) release, cardiac function and phosphorylated AKT and signal transducer and activator of transcription-3 (STAT-3) were measured. IPC significantly prevented the hearts from myocardial ischemia/reperfusion injury by decreasing infarct size, and CK and cTnT release in coronary effluent, and improving cardiac functional recovery in non-depressed rats. However, these cardioprotective effects of IPC were not observed in depressed rats. In addition, IPC had no effects on the phosphorylation of AKT and STAT-3 at reperfusion in depressed hearts, although it markedly increased the phosphorylation of AKT and STAT-3 at reperfusion in non-depressed hearts. In conclusion, these data indicate that cardioprotection by IPC is abolished during myocardial ischemia/reperfusion in depressed rats, and the underlying mechanisms are probably related to the impaired activation of AKT and STAT-3 at reperfusion.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychiatry, Anning Hospital of Tianjin City, No. 20 Yongping Lane, Dongli District, Tianjin, 300300, China
| | | | | | | | | |
Collapse
|
315
|
Postconditioning during primary percutaneous coronary intervention: A review and meta-analysis. Int J Cardiol 2010; 144:22-5. [DOI: 10.1016/j.ijcard.2009.03.118] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 03/11/2009] [Accepted: 03/26/2009] [Indexed: 11/20/2022]
|
316
|
BNP controls early load-dependent regulation of SERCA through calcineurin. Basic Res Cardiol 2010; 105:795-804. [PMID: 20711735 PMCID: PMC2965361 DOI: 10.1007/s00395-010-0115-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 07/20/2010] [Accepted: 08/03/2010] [Indexed: 02/06/2023]
Abstract
Heart failure is characterised by reduced expression of sarcoplasmic reticulum calcium-ATPase (SERCA) and increased expression of B-type natriuretic peptide (BNP). The present study was performed to investigate causality of this inverse relationship under in vivo conditions in the transversal aortic constriction mouse model (TAC). Left ventricular SERCA-mRNA expression was significantly upregulated in TAC by 32% after 6 h, but not different from sham after 24 h. Serum proANP and BNP levels were increased in TAC after 24 h (BNP +274%, p < 0.01; proANP +60%, p < 0.05), but only proANP levels were increased after 6 h (+182%, p < 0.01). cGMP levels were only increased 24 h after TAC (+307%, p < 0.01), but not 6 h after TAC. BNP infusion inhibited the increase in SERCA expression 6 h after TAC. In BNP-receptor-knockout animals (GC-A), the expression of SERCA was still significantly increased 24 h after TAC at the mRNA level by 35% (p < 0.05), as well as at the protein level by 25% (p < 0.05). MCIP expression as an indicator of calcineurin activity was regulated in parallel to SERCA after 6 and 24 h. MCIP-mRNA was increased by 333% 6 h after TAC, but not significantly different from sham after 24 h. In the GC-A-KO mice, MCIP-mRNA was significantly increased in TAC compared to WT after 24 h. In mice with BNP infusion, MCIP was significantly lower 6 h after TAC compared to control animals. In conclusion, mechanical load leads to an upregulation of SERCA expression. This is followed by upregulation of natriuretic peptides with subsequent suppression of SERCA upregulation. Elevated natriuretic peptides may suppress SERCA expression by inhibition of calcineurin activity via activation of GC-A.
Collapse
|
317
|
Abstract
Everyone encounters various stressors (causes of stress), such as psychological pressure, mental fluctuations, and physical burdens, in their everyday life. It is well accepted that the highest levels of perceived stress correlate with early onset of cardiovascular disease. Conversely, appropriate (mild to moderate) stressors, such as physical activity, have been shown to promote health. This bidirectional dose - response relationship of treatments that are beneficial at low levels but noxious at higher levels is referred to as "hormesis". In the fields of toxicology, pharmacology, radiation biology, and medicine, the significance of the biological effects of low-level exposure to various agents has attracted considerable attention. It is very important to understand how biological systems respond to low levels of stress and their implications within society. Aldehydes, the major endproducts of lipid peroxidation, have been implicated in the pathogenesis of oxidative stress-associated diseases. In addition to the pathogenic effect associated with oxidative stress, sublethal levels of aldehydes interact with signaling systems to upregulate the expression of genes to counteract the stressor challenge and to re-establish homeostasis. The present review article discusses current discoveries regarding the hormetic response to aldehyde and its clinical significance in cardioprotection.
Collapse
Affiliation(s)
- Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
318
|
Lønborg J, Treiman M, Engstrøm T. Ischemic postconditioning: a clinical perspective. Interv Cardiol 2010. [DOI: 10.2217/ica.10.52] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
319
|
Stoller DA, Fahrenbach JP, Chalupsky K, Tan BH, Aggarwal N, Metcalfe J, Hadhazy M, Shi NQ, Makielski JC, McNally EM. Cardiomyocyte sulfonylurea receptor 2-KATP channel mediates cardioprotection and ST segment elevation. Am J Physiol Heart Circ Physiol 2010; 299:H1100-8. [PMID: 20656890 DOI: 10.1152/ajpheart.00084.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sulfonylurea receptor-containing ATP-sensitive potassium (K(ATP)) channels have been implicated in cardioprotection, but the cell type and constitution of channels responsible for this protection have not been clear. Mice deleted for the first nucleotide binding region of sulfonylurea receptor 2 (SUR2) are referred to as SUR2 null since they lack full-length SUR2 and glibenclamide-responsive K(ATP) channels in cardiac, skeletal, and smooth muscle. As previously reported, SUR2 null mice develop electrocardiographic changes of ST segment elevation that were shown to correlate with coronary artery vasospasm. Here we restored expression of the cardiomyocyte SUR2-K(ATP) channel in SUR2 null mice by generating transgenic mice with ventricular cardiomyocyte-restricted expression of SUR2A. Introduction of the cardiomyocyte SUR2A transgene into the SUR2 null background restored functional cardiac K(ATP) channels. Hearts isolated from rescued mice, referred to as MLC2A, had significantly reduced infarct size (27 ± 3% of area at risk) compared with SUR2 null mice (36 ± 3% of area at risk). Compared with SUR2 null hearts, MLC2A hearts exhibited significantly improved cardiac function during the postischemia reperfusion period primarily because of preservation of low diastolic pressures. Additionally, restoration of cardiac SUR2-K(ATP) channels significantly reduced the degree and frequency of ST segment elevation episodes in MLC2A mice. Therefore, cardioprotective mechanisms both dependent and independent of SUR2-K(ATP) channels contribute to cardiac function.
Collapse
Affiliation(s)
- Douglas A Stoller
- Committee on Cellular and Molecular Physiology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
320
|
EMMPRIN activates multiple transcription factors in cardiomyocytes, and induces interleukin-18 expression via Rac1-dependent PI3K/Akt/IKK/NF-kappaB andMKK7/JNK/AP-1 signaling. J Mol Cell Cardiol 2010; 49:655-63. [PMID: 20538003 DOI: 10.1016/j.yjmcc.2010.05.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 05/17/2010] [Accepted: 05/20/2010] [Indexed: 12/31/2022]
Abstract
The transmembrane glycoprotein extracellular matrix metalloproteinase inducer (EMMPRIN), and the pleiotropic proinflammatory cytokine interleukin (IL)-18, play critical roles in myocardial remodeling, by inducing matrix degrading metalloproteinases (MMPs). Previously we showed that IL-18 induces EMMPRIN expression in cardiomyocytes via MyD88/IRAK4/TRAF6/JNK-dependent Sp1 activation. Here in reciprocal studies we demonstrate that EMMPRIN is a potent inducer of IL-18 transcription, protein expression and protein secretion in primary mouse cardiomyocytes. We show for the first time that EMMPRIN stimulates the activation of NF-kappaB, AP-1, CREB, and ATF-2 in cardiomyocytes, and induces IL-18 expression via Rac1-dependent PI3K/Akt/IKK/NF-kappaB and MKK7/JNK/AP-1 signaling. Moreover, EMMPRIN induces robust time-dependent induction of various MMP mRNAs. EMMPRIN also induces the mRNA of TIMPs 1 and 3, but in a delayed fashion. These results suggest that IL-18-induced EMMPRIN expression may favor net MMP expression and ECM destruction, and thus identify both as potential therapeutic targets in countering adverse myocardial remodeling.
Collapse
|
321
|
Chen G, Zhou X, Florea S, Qian J, Cai W, Zhang Z, Fan GC, Lorenz J, Hajjar RJ, Kranias EG. Expression of active protein phosphatase 1 inhibitor-1 attenuates chronic beta-agonist-induced cardiac apoptosis. Basic Res Cardiol 2010; 105:573-81. [PMID: 20512582 DOI: 10.1007/s00395-010-0106-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 05/17/2010] [Accepted: 05/19/2010] [Indexed: 11/28/2022]
Abstract
Cardiac apoptosis has been considered an important contributing factor to heart failure. Several subcellular mechanisms, including increased protein phosphatase 1 activity, have been suggested to induce apoptosis. Protein phosphatase 1 is regulated by an endogenous inhibitor-1 (I-1) that is activated upon phosphorylation at threonine 35 via protein kinase A. Here, we tested whether cardiac-specific overexpression of a constitutively active (T35D, AA 1-65) inhibitor-1 (I-1c), could also affect cardiac apoptosis and heart failure progression induced by prolonged beta-adrenergic stimulation. We found that either acute or chronic expression of I-1c reduced isoproterenol (ISO)-induced apoptosis assessed by nuclear condensation, TUNEL staining and DNA fragmentation. The beneficial effects of I-1c were associated with increased expression of the anti-apoptotic protein Bcl-2, decreased expression of the pro-apoptotic protein Bax and reduced levels of active caspases as well as increased activation of ERK. These findings suggest that mitochondrial signaling and ERK activation may be involved in the I-1c cardioprotective effects against apoptosis induced by prolonged beta-adrenergic stimulation.
Collapse
Affiliation(s)
- Guoli Chen
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, OH 45267-0575, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
Thielmann M, Kottenberg E, Boengler K, Raffelsieper C, Neuhaeuser M, Peters J, Jakob H, Heusch G. Remote ischemic preconditioning reduces myocardial injury after coronary artery bypass surgery with crystalloid cardioplegic arrest. Basic Res Cardiol 2010; 105:657-64. [PMID: 20495811 DOI: 10.1007/s00395-010-0104-5] [Citation(s) in RCA: 173] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 05/10/2010] [Accepted: 05/11/2010] [Indexed: 11/29/2022]
Abstract
Remote ischemic preconditioning (RIPC) with transient upper limb ischemia reduces myocardial injury in patients undergoing on-pump coronary artery bypass grafting (CABG) with cross-clamp fibrillation or blood cardioplegia for myocardial protection. Whether or not such protection is still operative when standard crystalloid cardioplegic arrest is used is uncertain. Fifty-three consecutive, non-diabetic patients with triple-vessel disease and 64 +/- 12 years of age (mean +/- SD), who underwent elective CABG surgery with crystalloid (Bretschneider) cardioplegic arrest, were allocated in a prospective, randomized, single-blinded protocol to receive either a RIPC protocol (3 cycles of 5 min transient left upper arm ischemia induced by inflating a blood pressure cuff to 200 mmHg with 5 min of reperfusion) or control, respectively, after induction of anesthesia. Cardiac troponin I (cTnI) concentration was measured preoperatively and over 72 h postoperatively, and the area under the curve (AUC) was calculated. Peak postoperative cTnI concentration was significantly reduced from 13.7 +/- 7.7 ng/mL in controls to 8.9 +/- 4.4 ng/mL in RIPC (P = 0.008). Mean cTnI concentration was significantly lower at 6, 12, 24, and 48 h after surgery (ANOVA; P < 0.0001) in the RIPC patients (N = 27) than in controls (N = 26), resulting in a 44.5% reduction of cTnI (AUC at 72 h). RIPC by repetitive inflation of a cuff around the left upper arm before surgery enhances myocardial protection in patients undergoing CABG surgery with antegrade cold crystalloid cardioplegia.
Collapse
Affiliation(s)
- Matthias Thielmann
- Department of Thoracic and Cardiovascular Surgery, West-German Heart Center Essen, University Hospital Essen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
323
|
Nitrative inactivation of thioredoxin-1 increases vulnerability of diabetic hearts to ischemia/reperfusion injury. J Mol Cell Cardiol 2010; 49:354-61. [PMID: 20497906 DOI: 10.1016/j.yjmcc.2010.05.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 05/04/2010] [Accepted: 05/05/2010] [Indexed: 01/08/2023]
Abstract
Hyperglycemia (HG) significantly increases mortality after myocardial infarction (MI) in patients with and without established diabetes. The specific underlying mechanism remains unknown. The present study attempted to determine whether nitrative inactivation of thioredoxin-1 (Trx-1) may contribute to the exaggerated myocardial ischemia/reperfusion (I/R) injury observed in the hyperglycemic condition. Diabetes was induced by multiple intraperitoneal injections of low-dose streptozotocin (STZ) in mice. After 30 min ischemia by slip-knot ligature of the left anterior descending coronary artery, the myocardium was reperfused for 3h after knot release (for apoptosis, Trx-1-activity, and -nitration determination) or 24h (for cardiac function and infarct size determination). At 10 min before reperfusion, diabetic mice were randomized to receive vehicle, EUK134 (a peroxynitrite scavenger), recombinant human Trx-1 (rhTrx-1), or SIN-1 (a peroxynitrite donor) nitrated Trx-1 (N-Trx-1) administration. Diabetes intensified I/R-induced myocardial injury, evidenced by further enlarged infarct size, increased apoptosis, and decreased cardiac function in diabetic mice. Trx-1 nitrative inactivation was elevated in the diabetic heart before I/R and was further amplified after I/R. Treatment with EUK134 or rhTrx-1, but not N-Trx-1, before reperfusion significantly reduced Trx-1 nitration, preserved Trx-1 activity, attenuated apoptosis, reduced infarct size, and improved cardiac function in diabetic mice. Taken together, our results demonstrated that HG increased cardiac vulnerability to I/R injury by enhancing nitrative inactivation of Trx-1, suggesting that blockade of Trx-1 nitration, or supplementation of exogenous rhTrx-1, might represent novel therapies to attenuate cardiac injury after MI in diabetic patients.
Collapse
|
324
|
Lim SY, Yellon DM, Hausenloy DJ. The neural and humoral pathways in remote limb ischemic preconditioning. Basic Res Cardiol 2010; 105:651-5. [DOI: 10.1007/s00395-010-0099-y] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 03/17/2010] [Accepted: 03/18/2010] [Indexed: 12/14/2022]
|
325
|
Ovize M, Baxter GF, Di Lisa F, Ferdinandy P, Garcia-Dorado D, Hausenloy DJ, Heusch G, Vinten-Johansen J, Yellon DM, Schulz R. Postconditioning and protection from reperfusion injury: where do we stand? Position paper from the Working Group of Cellular Biology of the Heart of the European Society of Cardiology. Cardiovasc Res 2010; 87:406-23. [PMID: 20448097 DOI: 10.1093/cvr/cvq129] [Citation(s) in RCA: 430] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Ischaemic postconditioning (brief periods of ischaemia alternating with brief periods of reflow applied at the onset of reperfusion following sustained ischaemia) effectively reduces myocardial infarct size in all species tested so far, including humans. Ischaemic postconditioning is a simple and safe manoeuvre, but because reperfusion injury is initiated within minutes of reflow, postconditioning must be applied at the onset of reperfusion. The mechanisms of protection by postconditioning include: formation and release of several autacoids and cytokines; maintained acidosis during early reperfusion; activation of protein kinases; preservation of mitochondrial function, most strikingly the attenuation of opening of the mitochondrial permeability transition pore (MPTP). Exogenous recruitment of some of the identified signalling steps can induce cardioprotection when applied at the time of reperfusion in animal experiments, but more recently cardioprotection was also observed in a proof-of-concept clinical trial. Indeed, studies in patients with an acute myocardial infarction showed a reduction of infarct size and improved left ventricular function when they underwent ischaemic postconditioning or pharmacological inhibition of MPTP opening during interventional reperfusion. Further animal studies and large-scale human studies are needed to determine whether patients with different co-morbidities and co-medications respond equally to protection by postconditioning. Also, our understanding of the underlying mechanisms must be improved to develop new therapeutic strategies to be applied at reperfusion with the ultimate aim of limiting the burden of ischaemic heart disease and potentially providing protection for other organs at risk of reperfusion injury, such as brain and kidney.
Collapse
Affiliation(s)
- Michel Ovize
- Service d'Explorations Fonctionnelles Cardiovasculaires and Inserm U886, Hospices Civils de Lyon, University of Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
326
|
|
327
|
Linares-Palomino J, Husainy MA, Lai VK, Dickenson JM, Galiñanes M. Selective blockade of protein kinase B protects the rat and human myocardium against ischaemic injury. J Physiol 2010; 588:2173-91. [PMID: 20403980 DOI: 10.1113/jphysiol.2010.190462] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein kinase B (PKB/Akt) plays a critical role in cell survival but the investigation of its involvement has been limited by the lack of specific pharmacological agents. In this study, using novel PKB inhibitors (VIII and XI), we investigated the role of PKB in cardioprotection of the rat and human myocardium, the location of PKB in relation to mitoK(ATP) channels and p38 mitogen-activated protein kinase (p38 MAPK), and whether the manipulation of PKB can overcome the unresponsiveness to protection of the diabetic myocardium. Myocardial slices from rat left ventricle and from the right atrial appendage of patients undergoing elective cardiac surgery were subjected to 90 min ischaemia/120 min reoxygenation at 37 degrees C. Tissue injury was assessed by creatine kinase (CK) released and determination of cell necrosis and apoptosis. The results showed that blockade of PKB activity caused significant reduction of CK release and cell death, a benefit that was as potent as ischaemic preconditioning and could be reproduced by blockade of phosphatidylinositol 3-kinase (PI-3K) with wortmannin and LY 294002. The protection was time dependent with maximal benefit seen when PKB and PI-3K were inhibited before ischaemia or during both ischaemia and reoxygenation. In addition, it was revealed that PKB is located downstream of mitoK(ATP) channels but upstream of p38 MAPK. PKB inhibition induced a similar degree of protection in the human and rat myocardium and, importantly, it reversed the unresponsiveness to protection of the diabetic myocardium. In conclusion, inhibition of PKB plays a critical role in protection of the mammalian myocardium and may represent a clinical target for the reduction of ischaemic injury.
Collapse
Affiliation(s)
- José Linares-Palomino
- Cardiac Surgery Unit, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | | | | | | | | |
Collapse
|
328
|
Shao ZH, Sharp WW, Wojcik KR, Li CQ, Han M, Chang WT, Ramachandran S, Li J, Hamann KJ, Vanden Hoek TL. Therapeutic hypothermia cardioprotection via Akt- and nitric oxide-mediated attenuation of mitochondrial oxidants. Am J Physiol Heart Circ Physiol 2010; 298:H2164-73. [PMID: 20382860 DOI: 10.1152/ajpheart.00994.2009] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Therapeutic hypothermia (TH) is a promising cardioprotective treatment for cardiac arrest and acute myocardial infarction, but its cytoprotective mechanisms remain unknown. In this study, we developed a murine cardiomyocyte model of ischemia-reperfusion injury to better determine the mechanisms of TH cardioprotection. We hypothesized that TH manipulates Akt, a survival kinase that mediates mitochondrial protection by modulating reactive oxygen species (ROS) and nitric oxide (NO) generation. Cardiomyocytes, isolated from 1- to 2-day-old C57BL6/J mice, were exposed to 90 min simulated ischemia and 3 h reperfusion. For TH, cells were cooled to 32 degrees C during the last 20 min of ischemia and the first hour of reperfusion. Cell viability was evaluated by propidium iodide and lactate dehydrogenase release. ROS production was measured by 6-carboxy-2',7'-dichlorodihydrofluorescein diacetate and mitochondrial membrane potential (DeltaPsim) by 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazoly-carbocyanine iodide (JC-1). Phospho (p)-Akt (Thr308), p-Akt (Ser473), and phosphorylated heat shock protein 27 (p-HSP27) (Ser82) were analyzed by Western blot analysis. TH attenuated reperfusion ROS generation, increased NO, maintained DeltaPsim, and decreased cell death [19.3 + or - 3.3% (n = 11) vs. 44.7 + or - 2.7% (n = 10), P < 0.001]. TH also increased p-Akt during ischemia before reperfusion. TH protection and attenuation of ROS were blocked by the inhibition of Akt and NO synthase but not by a cGMP inhibitor. HSP27, a regulator of Akt, also exhibited increased phosphorylation (Ser82) during ischemia with TH. We conclude that TH cardioprotection is mediated by enhanced Akt/HSP27 phosphorylation and enhanced NO generation, resulting in the attenuation of ROS generation and the maintenance of DeltaPsim following ischemia-reperfusion.
Collapse
Affiliation(s)
- Zuo-Hui Shao
- Section of Emergency Medicine, Department of Medicine, Emergency Resuscitation Center, University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
329
|
Rottlaender D, Boengler K, Wolny M, Michels G, Endres-Becker J, Motloch LJ, Schwaiger A, Buechert A, Schulz R, Heusch G, Hoppe UC. Connexin 43 acts as a cytoprotective mediator of signal transduction by stimulating mitochondrial KATP channels in mouse cardiomyocytes. J Clin Invest 2010; 120:1441-53. [PMID: 20364086 DOI: 10.1172/jci40927] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Potassium (K+) channels in the inner mitochondrial membrane influence cell function and survival. Increasing evidence indicates that multiple signaling pathways and pharmacological actions converge on mitochondrial ATP-sensitive K+ (mitoKATP) channels and PKC to confer cytoprotection against necrotic and apoptotic cell injury. However, the molecular structure of mitoKATP channels remains unresolved, and the mitochondrial phosphoprotein(s) that mediate cytoprotection by PKC remain to be determined. As mice deficient in the main sarcolemmal gap junction protein connexin 43 (Cx43) lack this cytoprotection, we set out to investigate a possible link among mitochondrial Cx43, mitoKATP channel function, and PKC activation. By patch-clamping the inner membrane of subsarcolemmal murine cardiac mitochondria, we found that genetic Cx43 deficiency, pharmacological connexin inhibition by carbenoxolone, and Cx43 blockade by the mimetic peptide 43GAP27 each substantially reduced diazoxide-mediated stimulation of mitoKATP channels. Suppression of mitochondrial Cx43 inhibited mitoKATP channel activation by PKC. MitoKATP channels of interfibrillar mitochondria, which do not contain any detectable Cx43, were insensitive to both PKC activation and diazoxide, further demonstrating the role of Cx43 in mitoKATP channel stimulation and the compartmentation of mitochondria in cell signaling. Our results define a role for mitochondrial Cx43 in protecting cardiac cells from death and provide a link between cytoprotective stimuli and mitoKATP channel opening, making Cx43 an attractive therapeutic target for protection against cell injury.
Collapse
Affiliation(s)
- Dennis Rottlaender
- Department of Internal Medicine III, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Ischemic preconditioning in the aging heart: from bench to bedside. Ageing Res Rev 2010; 9:153-62. [PMID: 19615470 DOI: 10.1016/j.arr.2009.07.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 07/05/2009] [Accepted: 07/06/2009] [Indexed: 11/23/2022]
Abstract
Coronary artery disease is the leading cause of death in industrialized countries for people older than 65 years of age. The reasons are still unclear. A reduction of endogenous mechanisms against ischemic insults has been proposed to explain this phenomenon. Cardiac ischemic preconditioning represents the most powerful endogenous protective mechanism against ischemia. Brief episodes of ischemia are able to protect the heart against a following more prolonged ischemic period. This protective mechanism seems to be reduced with aging both in experimental and clinical studies. Alterations of mediators release and/or intracellular pathways may be responsible for age-related ischemic preconditioning reduction. Opposite studies are questionable for the experimental model used, the timing of ischemic preconditioning, and the selection of elderly patients. Several pharmacological stimuli failed to mimic ischemic preconditioning in the aging heart but exercise training and caloric restriction separately, and more powerfully taken together, are able to completely preserve and/or restore the age-related reduction of ischemic preconditioning.
Collapse
|
331
|
Inhibition of mitochondrial permeability transition pore opening: the Holy Grail of cardioprotection. Basic Res Cardiol 2010; 105:151-4. [PMID: 20066536 DOI: 10.1007/s00395-009-0080-9] [Citation(s) in RCA: 237] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
332
|
Gorbe A, Giricz Z, Szunyog A, Csont T, Burley DS, Baxter GF, Ferdinandy P. Role of cGMP-PKG signaling in the protection of neonatal rat cardiac myocytes subjected to simulated ischemia/reoxygenation. Basic Res Cardiol 2010; 105:643-50. [PMID: 20349314 DOI: 10.1007/s00395-010-0097-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 03/11/2010] [Accepted: 03/15/2010] [Indexed: 12/14/2022]
Abstract
Nitric oxide (NO) and B-type natriuretic peptide (BNP) are protective against ischemia-reperfusion injury as they increase intracellular cGMP level via activation of soluble (sGC) or particulate guanylate cyclases (pGC), respectively. The aim of the present study was to examine if the cGMP-elevating mediators, NO and BNP, share a common downstream signaling pathway via cGMP-dependent protein kinase (PKG) in cardiac cytoprotection. Neonatal rat cardiac myocytes in vitro were subjected to 2.5 h simulated ischemia (SI) followed by 2 h reoxygenation. Cell viability was tested by trypan blue exclusion assay. PKG activity of cardiac myocytes was assessed by phospholamban (PLB) phosphorylation determined by western blot. Cell death was 34 +/- 2% after SI/reoxygenation injury in the control group. cGMP-inducing agents significantly decreased irreversible cell injury: the cGMP analog 8-bromo-cGMP (8-Br-cGMP, 10 nM) decreased it to 13 +/- 1% (p < 0.001), the direct NO-donor S-nitroso-N-acetylpenicillamine (SNAP, 1 microM) to 18 +/- 6% (p < 0.05) and BNP (10 nM) to 12 +/- 2% (p < 0.001), respectively. This protective effect was abolished by the selective PKG inhibitor KT-5823 (600 nM) in each case. As PLB is not a unique reporter for PKG activity since it is also phosphorylated by protein kinase A (PKA), we examined PLB phosphorylation in the presence of the PKA inhibitor KT-5720 (1 microM). The ratio of pPLB/PLB significantly increased after administration of both BNP and 8-Br-cGMP under ischemic conditions, which was abolished by the PKG inhibitor. This is the first demonstration that elevated cGMP produced either by the sGC activator SNAP or the pGC activator BNP exerts cytoprotective effects via a common downstream signaling pathway involving PKG activation.
Collapse
Affiliation(s)
- Aniko Gorbe
- Department of Biochemistry, University of Szeged, Hungary
| | | | | | | | | | | | | |
Collapse
|
333
|
You L, Li L, Xu Q, Ren J, Zhang F. Postconditioning reduces infarct size and cardiac myocyte apoptosis via the opioid receptor and JAK-STAT signaling pathway. Mol Biol Rep 2010; 38:437-43. [DOI: 10.1007/s11033-010-0126-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 03/17/2010] [Indexed: 01/05/2023]
|
334
|
Babiker FA, Lorenzen-Schmidt I, Mokelke E, Vanagt WY, Delhaas T, Waltenberger J, Cleutjens JP, Prinzen FW. Long-term protection and mechanism of pacing-induced postconditioning in the heart. Basic Res Cardiol 2010; 105:523-33. [PMID: 20336304 PMCID: PMC2862957 DOI: 10.1007/s00395-010-0095-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 02/18/2010] [Accepted: 03/13/2010] [Indexed: 11/30/2022]
Abstract
Brief periods of ventricular pacing during the early reperfusion phase (pacing-induced postconditioning, PPC) have been shown to reduce infarct size as measured after 2 h of reperfusion. In this study, we investigated (1) whether PPC leads to maintained reduction in infarct size, (2) whether abnormal mechanical load due to asynchronous activation is the trigger for PPC and (3) the signaling pathways that are involved in PPC. Rabbit hearts were subjected to 30 min of coronary occlusion in vivo, followed by 6 weeks of reperfusion. PPC consisted of ten 30-s intervals of left ventricular (LV) pacing, starting at reperfusion. PPC reduced infarct size (TTC staining) normalized to area at risk, from 49.0 ± 3.3% in control to 22.9 ± 5.7% in PPC rabbits. In isolated ejecting rabbit hearts, replacing LV pacing by biventricular pacing abolished the protective effect of PPC, whereas ten 30-s periods of high preload provided a protective effect similar to PPC. The protective effect of PPC was neither affected by the adenosine receptor blocker 8-SPT nor by the angiotensin II receptor blocker candesartan, but was abrogated by the cytoskeletal microtubule-disrupting agent colchicine. Blockers of the mitochondrial KATP channel (5HD), PKC (chelerythrine) and PI3-kinase (wortmannin) all abrogated the protection provided by PPC. In the in situ pig heart, PPC reduced infarct size from 35 ± 4 to 16 ± 12%, a protection which was abolished by the stretch-activated channel blocker gadolinium. No infarct size reduction was achieved if PPC application was delayed by 5 min or if only five pacing cycles were used. The present study indicates that (1) PPC permanently reduces myocardial injury, (2) abnormal mechanical loading is a more likely trigger for PPC than electrical stimulation or G-coupled receptor stimulation and (3) PPC may share downstream pathways with other modes of cardioprotection.
Collapse
Affiliation(s)
- Fawzi A Babiker
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
335
|
Lukowski R, Rybalkin SD, Loga F, Leiss V, Beavo JA, Hofmann F. Cardiac hypertrophy is not amplified by deletion of cGMP-dependent protein kinase I in cardiomyocytes. Proc Natl Acad Sci U S A 2010; 107:5646-51. [PMID: 20212138 PMCID: PMC2851748 DOI: 10.1073/pnas.1001360107] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It has been suggested that cGMP kinase I (cGKI) dampens cardiac hypertrophy. We have compared the effect of isoproterenol (ISO) and transverse aortic constriction (TAC) on hypertrophy in WT [control (CTR)] mice, total cGKI-KO mice, and cGKIbeta rescue mice (betaRM) lacking cGKI specifically in cardiomyocytes (CMs). Infusion of ISO did not change the expression of cGKI in the hearts of CTR mice or betaRM but raised the heart weight by approximately 20% in both. An identical hypertrophic growth response was measured in CMs from CTR mice and betaRM and in isolated adult CMs cultured with or without 1 muM ISO. In both genotypes, ISO infusion induced similar changes in the expression of hypertrophy-associated cardiac genes and significant elevation of serum atrial natriuretic peptide and total cardiac cGMP. No differences in cardiac hypertrophy were obtained by 7-day ISO infusion in 4- to 6-week-old conventional cGKI-KO and CTR mice. Furthermore, TAC-induced hypertrophy of CTR mice and betaRM was not different and did not result in changes of the cGMP-hydrolyzing phosphodiesterase activities in hypertropic hearts or CMs. These results strongly suggest that cardiac myocyte cGKI does not affect the development of heart hypertrophy induced by pressure overload or chronic ISO infusion.
Collapse
Affiliation(s)
- Robert Lukowski
- Forschergruppe 923 and
- Institut für Pharmakologie und Toxikologie, Technische Universität München, D-80802 Munich, Germany
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, D-72076 Tuebingen, Germany; and
| | - Sergei D. Rybalkin
- Forschergruppe 923 and
- Center for Integrated Protein Science Munich, Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280
| | - Florian Loga
- Forschergruppe 923 and
- Center for Integrated Protein Science Munich, Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
| | - Veronika Leiss
- Forschergruppe 923 and
- Center for Integrated Protein Science Munich, Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
| | - Joseph A. Beavo
- Forschergruppe 923 and
- Center for Integrated Protein Science Munich, Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280
| | - Franz Hofmann
- Forschergruppe 923 and
- Center for Integrated Protein Science Munich, Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
| |
Collapse
|
336
|
Heusch G. Adenosine and maximum coronary vasodilation in humans: myth and misconceptions in the assessment of coronary reserve. Basic Res Cardiol 2010; 105:1-5. [PMID: 19941145 DOI: 10.1007/s00395-009-0074-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
337
|
Fan WJ, van Vuuren D, Genade S, Lochner A. Kinases and phosphatases in ischaemic preconditioning: a re-evaluation. Basic Res Cardiol 2010; 105:495-511. [PMID: 20127248 DOI: 10.1007/s00395-010-0086-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 01/12/2010] [Accepted: 01/14/2010] [Indexed: 11/30/2022]
Abstract
Activation of several protein kinases occurs during myocardial ischaemia and during subsequent reperfusion. In contrast to the intensive investigation into the significance of kinase activation in cardioprotection, relatively little is known about the role of the phosphatases in this regard. The aim of this study was to re-evaluate the putative roles of PP1 and PP2A in ischaemia/reperfusion and in triggering ischaemic preconditioning. Isolated perfused working rat hearts were subjected to sustained global (15 or 20 min) or regional ischaemia (35 min), followed by reperfusion. Hearts were preconditioned using global ischaemia (1 x 5 or 3 x 5 min, alternated with 5 min reperfusion). To inhibit both PP1 and PP2A cantharidin (5 muM) was used. To inhibit PP2A only, okadaic acid (7.5 nM) was used. The drugs were administered during the preconditioning protocol, before onset of sustained ischaemia (pretreatment) or during reperfusion. Endpoints were mechanical recovery during reperfusion, infarct size and activation of PKB/Akt, p38 MAPK and ERK p42/p44, as determined by Western blot. Pretreatment of hearts with okadaic acid or cantharidin caused a significant reduction in mechanical recovery after 15 or 20 min global ischaemia. Administration of the drugs during an ischaemic preconditioning protocol abolished functional recovery during reperfusion and significantly increased infarct size. Administration of the drugs during reperfusion had no deleterious effects and increased functional recovery in 3 x PC hearts. To find an explanation for the differential effects of the inhibitors depending on the time of administration, hearts were freeze-clamped at different time points during the perfusion protocol. Administration of cantharidin before 5 min ischaemia activated all kinases. Subsequent reperfusion for 5 min without the drug maintained activation of the kinases until the onset of sustained ischaemia. Cantharidin given during preconditioning was associated with activation of p38MAPK and PKB/Akt during reperfusion after sustained ischaemia. However, administration of the drug during reperfusion only after sustained ischaemia caused activation of both PKB/Akt and ERK p42/p44. Phosphatase inhibition immediately prior to the onset of sustained ischaemia or during preconditioning abolishes protection during reperfusion, while inhibition of these enzymes during reperfusion either had no effect or enhanced the cardioprotective effects of preconditioning. It is proposed that inhibition of phosphatases during reperfusion may prolong the period of RISK activation and hence protect the heart.
Collapse
Affiliation(s)
- W J Fan
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Health Sciences, University of Stellenbosch, Tygerberg, Republic of South Africa
| | | | | | | |
Collapse
|
338
|
Heusch G, Kleinbongard P, Böse D, Levkau B, Haude M, Schulz R, Erbel R. Coronary microembolization: from bedside to bench and back to bedside. Circulation 2009; 120:1822-36. [PMID: 19884481 DOI: 10.1161/circulationaha.109.888784] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Coronary microembolization from the erosion or rupture of a vulnerable atherosclerotic plaque occurs spontaneously in acute coronary syndromes and iatrogenically during percutaneous coronary interventions. Typical consequences of coronary microembolization are microinfarcts with an inflammatory response, contractile dysfunction, and reduced coronary reserve. Apart from transient elevations of creatine kinase and troponin, microemboli can be visualized by intracoronary Doppler and the resulting microinfarcts by late-enhancement nuclear magnetic resonance. Statins, antiplatelet agents, and coronary vasodilators protect against microembolization and microinfarction when started before percutaneous coronary interventions. Distal protection devices can retrieve atherothrombotic debris and prevent its embolization into the microcirculation, but their effect on clinical outcome has been disappointing so far, except for saphenous vein bypass grafts. Devices for aspiration of thrombi and thrombus-derived vasoconstrictor, thrombogenic, and inflammatory substances, however, reduce thrombus burden, improve perfusion, and provide protection in patients with acute myocardial infarction.
Collapse
Affiliation(s)
- Gerd Heusch
- Institut für Pathophysiologie, Universitätsklinikum Essen, Essen, Germany.
| | | | | | | | | | | | | |
Collapse
|
339
|
Cardioselective nitric oxide synthase 3 gene transfer protects against myocardial reperfusion injury. Basic Res Cardiol 2009; 105:169-79. [PMID: 20020305 DOI: 10.1007/s00395-009-0077-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 11/23/2009] [Accepted: 12/01/2009] [Indexed: 10/20/2022]
Abstract
Nitric oxide modulates the severity of myocardial ischemia-reperfusion (I/R) injury. We investigated whether cardioselective nitric oxide synthase 3 (NOS3) gene transfer could confer myocardial protection against I/R injury in pigs and examined potential molecular mechanisms. I/R injury was induced by balloon occlusion of the left anterior descending artery for 45 min followed by 4 or 72 h reperfusion. Hemodynamic and pathological changes were measured in pigs in the absence (n = 11) or presence of prior intracoronary retroinfusion of human NOS3 (AdNOS3, 5 x 10(10) PFU, n = 13) or control vector (AdRR5, 5 x 10(10) PFU, n = 11). Retrograde NOS3 gene transfer selectively increased NOS3 expression and NO bioavailability in the area at risk (AAR) without changing endogenous NOS isoform expression. At 4 h R, LV systolic (dP/dt(max)) and diastolic (dP/dt(min)) function was better preserved in AdNOS3- than in AdRR5-injected pigs (2,539 +/- 165 vs. 1,829 +/- 156 mmHg/s, and -2,781 +/- 340 vs. -2,062 +/- 292 mmHg/s, respectively, P < 0.05 for both). Myocardial infarct size (% AAR) was significantly smaller in AdNOS3 than in control and AdRR5 and associated with a significantly greater reduction in cardiac myeloperoxidase activity, a marker of neutrophil infiltration. The latter effects were sustained at 72 h R in a subset of pigs (n = 7). In the AAR, intercellular endothelial adhesion molecule-1 expression and cardiomyocyte apoptosis were significantly lower in AdNOS3. In conclusion, single myocardial NOS3 retroinfusion attenuates I/R injury, and causes a sustained reduction in myocardial infarct size and inflammatory cell infiltration. Gene-based strategies to increase NO bioavailability may have therapeutic potential in myocardial I/R.
Collapse
|
340
|
Penna C, Tullio F, Moro F, Folino A, Merlino A, Pagliaro P. Effects of a protocol of ischemic postconditioning and/or captopril in hearts of normotensive and hypertensive rats. Basic Res Cardiol 2009; 105:181-92. [DOI: 10.1007/s00395-009-0075-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 11/27/2009] [Accepted: 11/30/2009] [Indexed: 12/19/2022]
|
341
|
Merkel MJ, Liu L, Cao Z, Packwood W, Young J, Alkayed NJ, Van Winkle DM. Inhibition of soluble epoxide hydrolase preserves cardiomyocytes: role of STAT3 signaling. Am J Physiol Heart Circ Physiol 2009; 298:H679-87. [PMID: 20008276 DOI: 10.1152/ajpheart.00533.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Soluble epoxide hydrolase (sEH) metabolizes epoxyeicosatrienoic acids (EETs), primarily 14,15-EET. EETs are derived from arachidonic acid via P-450 epoxygenases and are cardioprotective. We tested the hypothesis that sEH deficiency and pharmacological inhibition elicit tolerance to ischemia via EET-mediated STAT3 signaling in vitro and in vivo. In addition, the relevance of single nucleotide polymorphisms (SNPs) of EPHX2 (the gene encoding sEH) on tolerance to oxygen and glucose deprivation and reoxygenation and glucose repletion (OGD/RGR) was assessed in male C57BL\6J (WT) or sEH knockout (sEHKO) cardiomyocytes by using transactivator of transcription (TAT)-mediated transduction with sEH mutant proteins. Cell death and hydrolase activity was lower in Arg287Gln EPHX2 mutants vs. nontransduced controls. Excess 14,15-EET and SEH inhibition did not improve cell survival in Arg287Gln mutants. In WT cells, the putative EET receptor antagonist, 14,15-EEZE, abolished the effect of 14,15-EET and sEH inhibition. Cotreatment with 14,15-EET and SEH inhibition did not provide increased protection. In vitro, STAT3 inhibition blocked 14,15-EET cytoprotection, but not the effect of SEH inhibition. However, STAT3 small interfering RNA (siRNA) abolished cytoprotection by 14,15-EET and sEH inhibition, but cells pretreated with JAK2 siRNA remained protected. In vivo, STAT3 inhibition abolished 14,15-EET-mediated infarct size reduction. In summary, the Arg287Gln mutation is associated with improved tolerance against ischemia in vitro, and inhibition of sEH preserves cardiomyocyte viability following OGD/RGR via an EET-dependent mechanism. In vivo and in vitro, 14,15-EET-mediated protection is mediated in part by STAT3.
Collapse
Affiliation(s)
- Matthias J Merkel
- Anesthesiology Service, Veterans Affairs Medical Center, Mail Code: P3ANES, 3710 SW US Veterans Hospital Rd., Portland, OR 97239, USA
| | | | | | | | | | | | | |
Collapse
|
342
|
Tao L, Wang Y, Gao E, Zhang H, Yuan Y, Lau WB, Chan L, Koch WJ, Ma XL. Adiponectin: an indispensable molecule in rosiglitazone cardioprotection following myocardial infarction. Circ Res 2009; 106:409-17. [PMID: 19940263 DOI: 10.1161/circresaha.109.211797] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RATIONALE Patients treated with peroxisome proliferator-activated receptor (PPAR)-gamma agonist manifest favorable metabolic profiles associated with increased plasma adiponectin (APN). However, whether increased APN production as a result of PPAR-gamma agonist treatment is an epiphenomenon or is causatively related to the cardioprotective actions of PPAR-gamma remains unknown. OBJECTIVE To determine the role of APN in rosiglitazone (RSG) cardioprotection against ischemic heart injury. METHODS AND RESULTS Adult male wild-type (WT) and APN knockdown/knockout (APN(+ or -) and APN(- or -)) mice were treated with vehicle or RSG (20 mg/kg per day), and subjected to coronary artery ligation 3 days after beginning treatment. In WT mice, RSG (7 days) significantly increased adipocyte APN expression, elevated plasma APN levels (2.6-fold), reduced infarct size (17% reduction), decreased apoptosis (0.23 + or - 0.02% versus 0.47 + or - 0.04% TUNEL-positive in remote nonischemic area), attenuated oxidative stress (48.5% reduction), and improved cardiac function (P<0.01). RSG-induced APN production and cardioprotection were significantly blunted (P<0.05 versus WT) in APN(+ or -), and completely lost in APN(- or -) (P>0.05 versus vehicle-treated APN(- or -) mice). Moreover, treatment with RSG for up to 14 days significantly improved the postischemic survival rate of WT mice (P<0.05 versus vehicle group) but not APN knockdown/knockout mice. CONCLUSIONS The cardioprotective effects of PPAR-gamma agonists are critically dependent on its APN stimulatory action, suggesting that under pathological conditions where APN expression is impaired (such as advanced type 2 diabetes), the harmful cardiovascular effects of PPAR-gamma agonists may outweigh its cardioprotective benefits.
Collapse
Affiliation(s)
- Ling Tao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, 15 West Changle Rd, Xian, China, 710032.
| | | | | | | | | | | | | | | | | |
Collapse
|
343
|
Endo J, Sano M, Katayama T, Hishiki T, Shinmura K, Morizane S, Matsuhashi T, Katsumata Y, Zhang Y, Ito H, Nagahata Y, Marchitti S, Nishimaki K, Wolf AM, Nakanishi H, Hattori F, Vasiliou V, Adachi T, Ohsawa I, Taguchi R, Hirabayashi Y, Ohta S, Suematsu M, Ogawa S, Fukuda K. Metabolic Remodeling Induced by Mitochondrial Aldehyde Stress Stimulates Tolerance to Oxidative Stress in the Heart. Circ Res 2009; 105:1118-27. [DOI: 10.1161/circresaha.109.206607] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale
:
Aldehyde accumulation is regarded as a pathognomonic feature of oxidative stress–associated cardiovascular disease.
Objective
:
We investigated how the heart compensates for the accelerated accumulation of aldehydes.
Methods and Results
:
Aldehyde dehydrogenase 2 (ALDH2) has a major role in aldehyde detoxification in the mitochondria, a major source of aldehydes. Transgenic (Tg) mice carrying an
Aldh2
gene with a single nucleotide polymorphism (
Aldh2*2
) were developed. This polymorphism has a dominant-negative effect and the Tg mice exhibited impaired ALDH activity against a broad range of aldehydes. Despite a shift toward the oxidative state in mitochondrial matrices,
Aldh2*2
Tg hearts displayed normal left ventricular function by echocardiography and, because of metabolic remodeling, an unexpected tolerance to oxidative stress induced by ischemia/reperfusion injury. Mitochondrial aldehyde stress stimulated eukaryotic translation initiation factor 2α phosphorylation. Subsequent translational and transcriptional activation of activating transcription factor-4 promoted the expression of enzymes involved in amino acid biosynthesis and transport, ultimately providing precursor amino acids for glutathione biosynthesis. Intracellular glutathione levels were increased 1.37-fold in
Aldh2*2
Tg hearts compared with wild-type controls. Heterozygous knockout of
Atf4
blunted the increase in intracellular glutathione levels in
Aldh2*2
Tg hearts, thereby attenuating the oxidative stress–resistant phenotype. Furthermore, glycolysis and NADPH generation via the pentose phosphate pathway were activated in
Aldh2*2
Tg hearts. (NADPH is required for the recycling of oxidized glutathione.)
Conclusions
:
The findings of the present study indicate that mitochondrial aldehyde stress in the heart induces metabolic remodeling, leading to activation of the glutathione–redox cycle, which confers resistance against acute oxidative stress induced by ischemia/reperfusion.
Collapse
Affiliation(s)
- Jin Endo
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Motoaki Sano
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Takaharu Katayama
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Takako Hishiki
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Ken Shinmura
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Shintaro Morizane
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Tomohiro Matsuhashi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yoshinori Katsumata
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yan Zhang
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Hideyuki Ito
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yoshiko Nagahata
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Satori Marchitti
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Kiyomi Nishimaki
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Alexander Martin Wolf
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Hiroki Nakanishi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Fumiyuki Hattori
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Vasilis Vasiliou
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Takeshi Adachi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Ikuroh Ohsawa
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Ryo Taguchi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yoshio Hirabayashi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Shigeo Ohta
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Makoto Suematsu
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Satoshi Ogawa
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Keiichi Fukuda
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| |
Collapse
|
344
|
Ibanez B, Cimmino G, Prat-González S, Vilahur G, Hutter R, García MJ, Fuster V, Sanz J, Badimon L, Badimon JJ. The cardioprotection granted by metoprolol is restricted to its administration prior to coronary reperfusion. Int J Cardiol 2009; 147:428-32. [PMID: 19913314 DOI: 10.1016/j.ijcard.2009.09.551] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Revised: 08/17/2009] [Accepted: 09/01/2009] [Indexed: 12/18/2022]
Abstract
BACKGROUND Myocardial infarct size is a strong predictor of cardiovascular events. Intravenous metoprolol before coronary reperfusion has been shown to reduce infarct size; however, it is unknown whether oral metoprolol initiated early after reperfusion, as clinical guidelines recommend, is similarly cardioprotective. We compared the extent of myocardial salvage associated with intravenous pre-reperfusion-metoprolol administration in comparison with oral post-reperfusion-metoprolol or placebo. We also studied the effect on suspected markers of reperfusion injury. METHODS Thirty Yorkshire-pigs underwent a reperfused myocardial infarction, being randomized to pre-reperfusion-metoprolol, post-reperfusion-metoprolol or placebo. Cardiac magnetic resonance imaging was performed in eighteen pigs at day 3 for the quantification of salvaged myocardium. The amounts of at-risk and infarcted myocardium were quantified using T2-weighted and post-contrast delayed enhancement imaging, respectively. Twelve animals were sacrificed after 24h for reperfusion injury analysis. RESULTS The pre-reperfusion-metoprolol group had significantly larger salvaged myocardium than the post-reperfusion-metoprolol or the placebo groups (31 ± 4%, 13 ± 6%, and 7 ± 3% of myocardium at-risk respectively). Post-mortem analyses suggest lesser myocardial reperfusion injury in the pre-reperfusion-metoprolol in comparison with the other 2 groups (lower neutrophil infiltration, decreased myocardial apoptosis, and higher activation of the salvage-kinase phospho-Akt). Salvaged myocardium and reperfusion injury pair wise comparisons proved there were significant differences between the pre-reperfusion-metoprolol and the other 2 groups, but not among the latter two. CONCLUSIONS The intravenous administration of metoprolol before coronary reperfusion results in larger myocardial salvage than its oral administration initiated early after reperfusion. If confirmed in the clinical setting, the timing and route of β-blocker initiation could be revisited.
Collapse
Affiliation(s)
- Borja Ibanez
- Atherothrombosis Research Unit, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
345
|
Abete P, Cacciatore F, Testa G, Della-Morte D, Galizia G, Ferrara N, Rengo F. Clinical application of ischemic preconditioning in the elderly. Dose Response 2009; 8:34-40. [PMID: 20221286 DOI: 10.2203/dose-response.09-023.abete] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
A mild stress such as brief ischemic episodes may protect the heart from a successive and more prolonged myocardial ischemia (ischemic preconditioning). This phenomenon is considered a typical "hormetic mechanism" by which the heart is immunized from pathological insults such as myocardial ischemia. This mechanism is reduced with aging and it may be restored and/or preserved by drugs such as adenosine or nicorandil, a mitochondrial K(ATP) channels, and lifestyle interventions such as physical activity and/or hypocaloric diet. Moreover, since the mechanisms involved in cardiac ischemic preconditioning have been established basic and clinical investigators are encouraged to test several drug in well-controlled animal and human studies in order to prevent and/or restore the age-related reduction of ischemic preconditioning.
Collapse
Affiliation(s)
- Pasquale Abete
- Dipartimento di Medicina Clinica, Scienze Cardiovascolari ed Immunologiche, Cattedra di Geriatria, Università degli Studi di Napoli Federico II, Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
346
|
Postconditioning with levosimendan reduces the infarct size involving the PI3K pathway and KATP-channel activation but is independent of PDE-III inhibition. Basic Res Cardiol 2009; 105:155-67. [DOI: 10.1007/s00395-009-0064-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 09/15/2009] [Accepted: 09/21/2009] [Indexed: 10/20/2022]
|
347
|
Létienne R, Bel L, Bessac AM, Vacher B, Le Grand B. Myocardial protection by F 15845, a persistent sodium current blocker, in an ischemia-reperfusion model in the pig. Eur J Pharmacol 2009; 624:16-22. [PMID: 19778535 DOI: 10.1016/j.ejphar.2009.09.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 09/10/2009] [Accepted: 09/11/2009] [Indexed: 12/19/2022]
Abstract
The specific persistent sodium current blocker F 15845 was tested in two myocardial ischemia-reperfusion models in the pig in order to evaluate its cardioprotective effects. In the first protocol, the left circumflex coronary artery was ligated for 60-min and then reperfused for 48-h. F 15845 (2.5+2.5 and 5+5mg/kg) was administered by i.v. infusion, starting before ischemia to the beginning of reperfusion. The second protocol attempted to evaluate F 15845 (5+5mg/kg) response in a more pathological state of the heart. To this end, a non necrotic ligation of the left circumflex coronary artery was applied for 15 min one week before the actual 60 min occlusion. For both protocols, infarct size was determined at the end of the reperfusion period and was assessed by histochemistry (tetrazolium staining). Plasma levels of biochemical markers (myoglobin and troponin I) were also evaluated. In protocol 1, F 15845 significantly reduced the infarct size by 27+/-3 and 43+/-5% at 2.5+2.5 and 5+5mg/kg, respectively. At 5+5mg/kg, F 15845 decreased plasma levels of myoglobin and cardiac troponin I. In protocol 2, F 15845 (5+5mg/kg) significantly reduced myocardial infarct size by 54+/-15% and lowered the plasma myoglobin and troponin I levels relative to vehicle-treated animals. In conclusion, the highly effective persistent sodium current blocker F 15845 exerts remarkable cardioprotective activities. It reduces both myocardial infarct size and the release of biochemical markers in healthy pigs as well in pigs previously exposed to an ischemic episode.
Collapse
Affiliation(s)
- Robert Létienne
- Centre de Recherche Pierre Fabre, 17 Avenue Jean Moulin, 81106 Castres Cedex, France
| | | | | | | | | |
Collapse
|
348
|
Iliodromitis EK, Downey JM, Heusch G, Kremastinos DT. What is the optimal postconditioning algorithm? J Cardiovasc Pharmacol Ther 2009; 14:269-73. [PMID: 19741111 DOI: 10.1177/1074248409344328] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ischemic postconditioning has emerged as a clinically feasible intervention for limiting infarction in the setting of percutaneous intervention. In ischemic postconditioning, a number of cycles of a brief period of reperfusion followed by a brief period of occlusion are applied immediately upon reperfusion of the ischemic heart. Although ischemic postconditioning is protective in both animals and man, the animal studies reveal that the algorithm used in selecting the duration of the occlusion and reperfusion periods is critical to the degree of protection realized and it varies with species. The question then arises what is the best algorithm for man? The available animal and clinical data are examined in an attempt to shed light on this perplexing problem.
Collapse
Affiliation(s)
- Efstathios K Iliodromitis
- 2nd University Department of Cardiology, Medical School, Attikon General Hospital, University of Athens, Athens, Greece
| | | | | | | |
Collapse
|
349
|
Miro-Casas E, Ruiz-Meana M, Agullo E, Stahlhofen S, Rodríguez-Sinovas A, Cabestrero A, Jorge I, Torre I, Vazquez J, Boengler K, Schulz R, Heusch G, Garcia-Dorado D. Connexin43 in cardiomyocyte mitochondria contributes to mitochondrial potassium uptake. Cardiovasc Res 2009; 83:747-56. [PMID: 19460776 DOI: 10.1093/cvr/cvp157] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
AIMS Connexin43 is present at the inner membrane of cardiomyocyte mitochondria (mCx43), but its function remains unknown. METHODS AND RESULTS In this study we verified the presence of mCx43 by a mass spectrometry-based proteomic approach in purified mitochondrial preparations from mouse myocardium and determined by western blot analysis that the C-terminus of mCx43 is oriented towards the intermembrane space. Cross-linking studies with dimethylsuberimidate indicated the presence of Cx43 hexamers in mitochondrial membranes. The contribution of Cx43 to both mitochondrial dye uptake and K(+) flux was assessed in wild-type mice using hemichannel blockers and Cx43KI32 mice in which Cx43 had been replaced by Cx32. Uptake of the Cx43 hemichannel-permeant dye Lucifer Yellow was reduced in mitochondria from wild-type mice by two hemichannel blockers (carbenoxolone and heptanol) and in Cx43KI32 compared with wild-type mice. Mitochondrial K(+) influx (PBFI fluorescence) was decreased in digitonin-permeabilized cardiomyocytes from Cx32 mutants compared with wild-type mice, and addition of the Cx43 hemichannel blocker 18alpha-glycyrrhetinic acid had an inhibitory effect on mitochondrial K(+) influx in wild-type cardiomyocytes, but not in cardiomyocytes from Cx32 mutants. CONCLUSION These results indicate that mCx43 contributes to mitochondrial K(+) flux in cardiomyocytes, potentially by forming hemichannel-like structures.
Collapse
Affiliation(s)
- Elisabet Miro-Casas
- Servicio de Cardiología, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119, Barcelona 08035, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
350
|
Heusch G. A BEAUTIFUL lesson--ivabradine protects from ischaemia, but not from heart failure: through heart rate reduction or more? Eur Heart J 2009; 30:2300-1. [PMID: 19720636 DOI: 10.1093/eurheartj/ehp360] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Gerd Heusch
- Institut für Pathophysiologie, Universitätsklinikum Essen, D-45122 Essen, Germany.
| |
Collapse
|