1
|
Yusof NLM, Yellon DM, Davidson SM. Novel Selective Cardiac Myosin-Targeted Inhibitors Alleviate Myocardial Ischaemia-Reperfusion Injury. Cardiovasc Drugs Ther 2025:10.1007/s10557-024-07663-0. [PMID: 39754660 DOI: 10.1007/s10557-024-07663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/06/2025]
Abstract
PURPOSE Reperfusion of the ischaemic heart is essential to limit myocardial infarction. However, reperfusion can cause cardiomyocyte hypercontracture. Recently, cardiac myosin-targeted inhibitors (CMIs), such as Mavacamten (MYK-461) and Aficamten (CK-274), have been developed to treat patients with cardiac hypercontractility. These CMIs are well tolerated and safe in clinical trials. We hypothesised that, by limiting hypercontraction, CMIs may reduce hypercontracture and protect hearts in the setting of ischaemia and reperfusion (IR). METHODS We investigated the ability of MYK-461 and CK-274 to inhibit hypercontracture of adult rat cardiomyocytes (ARVC) in vitro following ATP depletion. A suitable dose of CMIs for subsequent in vivo IR studies was identified using cardiac echocardiography of healthy male Sprague Dawley rats. Rats were anaesthetized and subject to coronary artery ligation for 30 min followed by 2 h of reperfusion. Prior to reperfusion, CMI or vehicle was administered intraperitoneally. Ischaemic preconditioning (IPC) was used as a positive control group. Infarct size was assessed by tetrazolium chloride staining and extent of hypercontracture was assessed by histological staining. RESULTS Treatment with CMIs inhibited ARVC hypercontracture in vitro. MYK-461 (2 mg/kg) and CK-274 (0.5 mg/kg to 2 mg/kg) significantly reduced infarct size vs. vehicle. IR caused extensive contraction band necrosis, which was reduced significantly by IPC but not by CMIs, likely due to assay limitations. GDC-0326, an inhibitor of PI3Kα, abrogated CK-274-mediated protection following IR injury. GDC-0326 reduced phosphorylation of AKT when administered together with CK-274. CONCLUSION This study identifies CMIs as novel cardioprotective agents in the setting of IR injury.
Collapse
Affiliation(s)
- Nur Liyana Mohammed Yusof
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
2
|
Yang ZJ, Zhang WF, Jin QQ, Wu ZR, Du YY, Shi H, Qu ZS, Han XJ, Jiang LP. Lactate Contributes to Remote Ischemic Preconditioning-Mediated Protection Against Myocardial Ischemia Reperfusion Injury by Facilitating Autophagy via the AMP-Activated Protein Kinase-Mammalian Target of Rapamycin-Transcription Factor EB-Connexin 43 Axis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1857-1878. [PMID: 39069170 DOI: 10.1016/j.ajpath.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/06/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Remote ischemic preconditioning (RIPC) exerts a protective role on myocardial ischemia/reperfusion (I/R) injury by the release of various humoral factors. Lactate is a common metabolite in ischemic tissues. Nevertheless, little is known about the role lactate plays in myocardial I/R injury and its underlying mechanism. This investigation revealed that RIPC elevated the level of lactate in blood and myocardium. Furthermore, AZD3965, a selective monocarboxylate transporter 1 inhibitor, and 2-deoxy-d-glucose, a glycolysis inhibitor, mitigated the effects of RIPC-induced elevated lactate in the myocardium and prevented RIPC against myocardial I/R injury. In an in vitro hypoxia/reoxygenation model, lactate markedly mitigated hypoxia/reoxygenation-induced cell damage in H9c2 cells. Further studies suggested that lactate contributed to RIPC, rescuing I/R-induced autophagy deficiency by promoting transcription factor EB (TFEB) translocation to the nucleus through activating the AMP-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR) pathway without influencing the phosphatidylinositol 3-kinase-Akt pathway, thus reducing cardiomyocyte damage. Interestingly, lactate up-regulated the mRNA and protein expression of connexin 43 (CX43) by facilitating the binding of TFEB to CX43 promoter in the myocardium. Functionally, silencing of TFEB attenuated the protective effect of lactate on cell damage, which was reversed by overexpression of CX43. Further mechanistic studies suggested that lactate facilitated CX43-regulated autophagy via the AMPK-mTOR-TFEB signaling pathway. Collectively, this research demonstrates that RIPC protects against myocardial I/R injury through lactate-mediated myocardial autophagy via the AMPK-mTOR-TFEB-CX43 axis.
Collapse
Affiliation(s)
- Zhang-Jian Yang
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China; Department of Pharmacy, 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wei-Fang Zhang
- Department of Pharmacy, 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qing-Qing Jin
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhi-Rong Wu
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yun-Yan Du
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hao Shi
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhen-Sheng Qu
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| | - Li-Ping Jiang
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
3
|
Kobayashi Y, Li J, Parker M, Wang J, Nagy A, Fan CPS, Runeckles K, Okumura M, Kadowaki S, Honjo O. Impact of Hemoglobin Level in Ex Vivo Heart Perfusion on Donation After Circulatory Death Hearts: A Juvenile Porcine Experimental Model. Transplantation 2024; 108:1922-1930. [PMID: 39167562 DOI: 10.1097/tp.0000000000004954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
BACKGROUND Ex vivo heart perfusion (EVHP) of donation after circulatory death (DCD) hearts has become an effective strategy in adults; however, the small circulating volume in pediatrics poses the challenge of a low-hemoglobin (Hb) perfusate. We aimed to determine the impact of perfusate Hb levels during EVHP on DCD hearts using a juvenile porcine model. METHODS Sixteen DCD piglet hearts (11-14 kg) were reperfused for 4 h in unloaded mode followed by working mode. Metabolism, cardiac function, and cell damage were compared between the low-Hb (Hb, 5.0-5.9 g/dL; n = 8) and control (Hb, 7.5-8.4 g/dL; n = 8) groups. Between-group differences were evaluated using 2-sample t -tests or Fisher's Exact tests. RESULTS During unloaded mode, the low-Hb group showed lower myocardial oxygen consumption ( P < 0.001), a higher arterial lactate level ( P = 0.001), and worse systolic ventricular function ( P < 0.001). During working mode, the low-Hb group had a lower cardiac output (mean, 71% versus 106% of normal cardiac output, P = 0.010) and a higher arterial lactate level ( P = 0.031). Adjusted cardiac troponin-I ( P = 0.112) did not differ between the groups. Morphological myocyte injury in the left ventricle was more severe in the low-Hb group ( P = 0.028). CONCLUSIONS Low-Hb perfusate with inadequate oxygen delivery induced anaerobic metabolism, resulting in suboptimal DCD heart recovery and declined cardiac function. Arranging an optimal perfusate is crucial to organ protection, and further endeavors to refine the priming volume of EVHP or the transfusion strategy are required.
Collapse
Affiliation(s)
- Yasuyuki Kobayashi
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Jing Li
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Marlee Parker
- Division of Perfusion Services, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jian Wang
- Division of Perfusion Services, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anita Nagy
- Division of Pathology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Chun-Po Steve Fan
- Ted Rogers Computational Program, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kyle Runeckles
- Ted Rogers Computational Program, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Michiru Okumura
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Sachiko Kadowaki
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Osami Honjo
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Bannerman D, Gil de Gomez SP, Wu Q, Fernandes I, Zhao Y, Wagner KT, Okhovatian S, Landau S, Raftian N, Bodenstein DF, Wang Y, Nash TR, Vunjak-Novakovic G, Keller G, Epelman S, Radisic M. Heart-on-a-Chip Model of Epicardial-Myocardial Interaction in Ischemia Reperfusion Injury. Adv Healthc Mater 2024; 13:e2302642. [PMID: 38683053 PMCID: PMC11338737 DOI: 10.1002/adhm.202302642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 03/22/2024] [Indexed: 05/01/2024]
Abstract
Epicardial cells (EPIs) form the outer layer of the heart and play an important role in development and disease. Current heart-on-a-chip platforms still do not fully mimic the native cardiac environment due to the absence of relevant cell types, such as EPIs. Here, using the Biowire II platform, engineered cardiac tissues with an epicardial outer layer and inner myocardial structure are constructed, and an image analysis approach is developed to track the EPI cell migration in a beating myocardial environment. Functional properties of EPI cardiac tissues improve over two weeks in culture. In conditions mimicking ischemia reperfusion injury (IRI), the EPI cardiac tissues experience less cell death and a lower impact on functional properties. EPI cell coverage is significantly reduced and more diffuse under normoxic conditions compared to the post-IRI conditions. Upon IRI, migration of EPI cells into the cardiac tissue interior is observed, with contributions to alpha smooth muscle actin positive cell population. Altogether, a novel heart-on-a-chip model is designed to incorporate EPIs through a formation process that mimics cardiac development, and this work demonstrates that EPI cardiac tissues respond to injury differently than epicardium-free controls, highlighting the importance of including EPIs in heart-on-a-chip constructs that aim to accurately mimic the cardiac environment.
Collapse
Affiliation(s)
- Dawn Bannerman
- Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Simon Pascual Gil de Gomez
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Qinghua Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Ian Fernandes
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Karl T. Wagner
- Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Naimeh Raftian
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - David F. Bodenstein
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
- Department of Toxicology, University of Toronto, Toronto, ON, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| | - Trevor R. Nash
- Department of Medicine, Columbia University, New York, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Gordon Keller
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Slava Epelman
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
- Division of Cardiology, University Health Network, Peter Munk Cardiac Centre
| | - Milica Radisic
- Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Health Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
5
|
Li T, Li Y, Zeng Y, Zhou X, Zhang S, Ren Y. Construction of preclinical evidence for propofol in the treatment of reperfusion injury after acute myocardial infarction: A systematic review and meta-analysis. Biomed Pharmacother 2024; 174:116629. [PMID: 38640712 DOI: 10.1016/j.biopha.2024.116629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2024] Open
Abstract
Propofol, a commonly used intravenous anesthetic, has demonstrated potential in protecting against myocardial ischemia/reperfusion injury (MIRI) based on preclinical animal studies. However, the clinical benefits of propofol in this context are subject to debate. We conducted a systematic search across eight databases to identify all relevant animal studies investigating the preventive effects of propofol on MIRI until October 30, 2023. We assessed the methodological quality of the included studies using SYRCLE's bias risk tool. Statistical analysis was performed using STATA 15.1. The primary outcome measures analyzed in this study were myocardial infarct size (IS) and myocardial injury biomarkers. This study presents a comprehensive analysis of 48 relevant animal studies investigating propofol's preventive effects on MIRI. Propofol administration demonstrated a reduction in myocardial IS and decreased levels of myocardial injury biomarkers (CK-MB, LDH, cTnI). Moreover, propofol improved myocardial function parameters (+dp/dtmax, -dP/dtmax, LVEF, LVFS), exhibited favorable effects on inflammatory markers (IL-6, TNF-α) and oxidative stress markers (SOD, MDA), and reduced myocardial cell apoptotic index (AI). These findings suggest propofol exerts cardioprotective effects by reducing myocardial injury, decreasing infarct size, and improving heart function. However, the absence of animal models that accurately represent comorbidities such as aging and hypertension, as well as inconsistent administration methods that align with clinical practice, may hinder its clinical translation. Further robust investigations are required to validate these findings, elucidate the underlying mechanisms of propofol, and facilitate its potential translation into clinical practice.
Collapse
Affiliation(s)
- Tao Li
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanwei Li
- Cardiology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiwei Zeng
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Zhou
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Su Zhang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulan Ren
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
6
|
Wang X, Ren L, Chen S, Tao Y, Zhao D, Wu C. Long non-coding RNA MIR4435-2HG/microRNA-125a-5p axis is involved in myocardial ischemic injuries. Bioengineered 2022; 13:10707-10720. [PMID: 35475469 PMCID: PMC9208505 DOI: 10.1080/21655979.2022.2051259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
This study aimed to investigate whether and how long non-coding RNA (lncRNA) MIR4435-2 host gene (MIR4435-2HG) involved in acute myocardial ischemia/reperfusion (I/R). Blood samples were collected from acute myocardial infarction (AMI) patients to detect MIR4435-2HG expression. In vivo myocardial I/R mice model and in vitro H2O2-induced oxidative stress model were established. Echocardiography, TUNEL assay and lactate dehydrogenase (LDH) detection were performed to assess heart infarction and myocardium apoptosis. Relationship among microRNA-125a-5p (miR-125a-5p), MIR4435-2HG and Mitochondrial fission protein 1 (MTFP1) was predicted by Targetscan and verified by luciferase reporter assay. MIR4435-2HG was notably upregulated in AMI patients, myocardial I/R mice and H2O2-treated cells. Knockdown of MIR4435-2HG notably alleviated infraction volume, ejection fraction (EF) and fractional shortening (FS) levels, cell apoptosis portion and pro-apoptotic cleaved-caspase-3 and Cyt c expression caused by myocardial I/R and oxidative stress, as well as improved cardiomyocytes viability. Transfection with miR-125a-5p alleviated MIR4435-2HG-caused cardiomyocytes apoptosis during oxidative stress. MiR-125a-5p overexpression decreased luciferase activity of the wild-type MIR4435-2HG compared with the mutated MIR4435-2HG. The expression levels of MTFP1 were elevated in myocardium from MI mice model and H2O2-treated AC16 cardiomyocytes. In addition, miR-125a-5p overexpression inhibited MTFP1 expression, and could stimulate the wild-type MTFP1 promoter luciferase activity but not the mutated one. Our findings revealed the role of MIR4435-2HG in MI-induced myocardium injury and cardiomyocytes apoptosis, disclosed a novel MIR4435-2HG/miR-125a-5p regulatory axis during myocardial I/R, and thus identified a potential target for the therapy of myocardial IR injury.
Collapse
Affiliation(s)
- Xiuling Wang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Lina Ren
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Shuai Chen
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Yanli Tao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Dandan Zhao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Chunwei Wu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| |
Collapse
|
7
|
Ma H, Hao J, Liu H, Yin J, Qiang M, Liu M, He S, Zeng D, Liu X, Lian C, Gao Y. Peoniflorin Preconditioning Protects Against Myocardial Ischemia/Reperfusion Injury Through Inhibiting Myocardial Apoptosis: RISK Pathway Involved. Appl Biochem Biotechnol 2022; 194:1149-1165. [PMID: 34596828 DOI: 10.1007/s12010-021-03680-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/08/2021] [Indexed: 11/28/2022]
Abstract
Preconditioning with Peoniflorin, a component of traditional Chinese prescriptions, was proposed to be a potential strategy for cardioprotection against ischemia/reperfusion (I/R) injury. However, the cardioprotective effect of Peoniflorin preconditioning has not been thoroughly confirmed, and the underlying mechanism remains unclear. Here, we examined the cardioprotective effect and its mechanism of Peoniflorin preconditioning against myocardial I/R injury. Rats were subjected to 30 min of transient ischemia followed by 2 h of reperfusion with or without Peoniflorin (100 mg/kg) prior to reperfusion. Peoniflorin preconditioning significantly limited myocardial infarct size and reperfusion arrhythmias, as well as obviously attenuated the histomorphological and micromorphological damages induced by I/R injury. The reduced myocardial injury was also associated with the anti-apoptotic effect of Peoniflorin, as evidence by decreased TUNEL-positive cells, upregulation of BCL-2 expression, and downregulation of Bax and caspase-3 expression. In an effort to evaluate the mechanism responsible for the observed cardioprotective and anti-apoptotic effect, Western blot of phosphorylated protein was performed after 20 min of reperfusion. Results showed that Peoniflorin preconditioning activated both the Akt and ERK1/2 arm of the reperfusion injury salvage kinase (RISK) pathway. To further confirm this mechanism, the PI3K signaling inhibitor LY294002 and ERK1/2 signaling inhibitor PD98059 were administered in vivo. The cardioprotective and anti-apoptotic effects of Peoniflorin preconditioning were diminished but not abolished by pretreatment with LY294002 or PD98059. Taken together, these results indicate that Peoniflorin preconditioning protects the myocardial against I/R injury and inhibits myocardial apoptosis via the activation of the RISK pathway, highlighting the potential therapeutic effects of Peoniflorin on reducing myocardial I/R injury.
Collapse
Affiliation(s)
- Hongen Ma
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Jiping Hao
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Huihui Liu
- Medical College of Yan'an University, No. 38 Guanghua RoadBaota District, Yan'an, 716000, Shaanxi, China
| | - Jia Yin
- Medical College of Yan'an University, No. 38 Guanghua RoadBaota District, Yan'an, 716000, Shaanxi, China
| | - Mingmin Qiang
- Medical College of Yan'an University, No. 38 Guanghua RoadBaota District, Yan'an, 716000, Shaanxi, China
| | - Meilin Liu
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Shaohui He
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi RoadBaqiao District, Xi'an, 710000, Shaanxi, China
| | - Xiongtao Liu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi RoadBaqiao District, Xi'an, 710000, Shaanxi, China
| | - Cheng Lian
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi RoadBaqiao District, Xi'an, 710000, Shaanxi, China
| | - Yuqin Gao
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
8
|
Fischesser DM, Bo B, Benton RP, Su H, Jahanpanah N, Haworth KJ. Controlling Reperfusion Injury With Controlled Reperfusion: Historical Perspectives and New Paradigms. J Cardiovasc Pharmacol Ther 2021; 26:504-523. [PMID: 34534022 DOI: 10.1177/10742484211046674] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiac reperfusion injury is a well-established outcome following treatment of acute myocardial infarction and other types of ischemic heart conditions. Numerous cardioprotection protocols and therapies have been pursued with success in pre-clinical models. Unfortunately, there has been lack of successful large-scale clinical translation, perhaps in part due to the multiple pathways that reperfusion can contribute to cell death. The search continues for new cardioprotection protocols based on what has been learned from past results. One class of cardioprotection protocols that remain under active investigation is that of controlled reperfusion. This class consists of those approaches that modify, in a controlled manner, the content of the reperfusate or the mechanical properties of the reperfusate (e.g., pressure and flow). This review article first provides a basic overview of the primary pathways to cell death that have the potential to be addressed by various forms of controlled reperfusion, including no-reflow phenomenon, ion imbalances (particularly calcium overload), and oxidative stress. Descriptions of various controlled reperfusion approaches are described, along with summaries of both mechanistic and outcome-oriented studies at the pre-clinical and clinical phases. This review will constrain itself to approaches that modify endogenously-occurring blood components. These approaches include ischemic postconditioning, gentle reperfusion, controlled hypoxic reperfusion, controlled hyperoxic reperfusion, controlled acidotic reperfusion, and controlled ionic reperfusion. This review concludes with a discussion of the limitations of past approaches and how they point to potential directions of investigation for the future.
Collapse
Affiliation(s)
- Demetria M Fischesser
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Bin Bo
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Rachel P Benton
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Haili Su
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Newsha Jahanpanah
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Kevin J Haworth
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
9
|
Zhao L, Jiang S, Wu N, Shi E, Yang L, Li Q. MiR-17-5p-mediated endoplasmic reticulum stress promotes acute myocardial ischemia injury through targeting Tsg101. Cell Stress Chaperones 2021; 26:77-90. [PMID: 32895884 PMCID: PMC7736418 DOI: 10.1007/s12192-020-01157-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/18/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death globally, among which acute myocardial infarction (AMI) frequently occurs in the heart and proceeds from myocardium ischemia and endoplasmic reticulum (ER) stress-induced cell death. Numerous studies on miRNAs indicated their potential as diagnostic biomarkers and treatment targets for heart diseases. Our study investigated the role of miR-17-5p and its regulatory mechanisms during AMI. Echocardiography, MTT, flow cytometry assay, evaluation of caspase-3 and lactate dehydrogenase (LDH) activity were conducted to assess cell viability, apoptosis in an MI/R mice model, and an H2O2-induced H9c2 hypoxia cell model, respectively. The expression levels of ER stress response-related biomarkers were detected using qRT-PCR, IHC, and western blotting assays. The binding site of miR-17-5p on Tsg101 mRNA was determined by bioinformatic prediction and luciferase reporter assay. The expression levels of miR-17-5p were notably elevated in MI/R mice and hypoxia cell models, accompanied by enhanced cell apoptosis. Inhibition of miR-17-5p led to decreased apoptosis related to ER stress response in the hypoxia model, which could be counteracted by knockdown of Tsg101 (tumor susceptibility gene 101). Transfection with miR-17-5p mimics downregulated the expression of Tsg101 in H9c2 cells. Luciferase assay demonstrated the binding between miR-17-5p and Tsg101. Moreover, 4-PBA, the inhibitor of the ER stress response, abolished shTsg101 elevated apoptosis in hypoxic H9c2 cells. Our findings investigated the pro-apoptotic role of miR-17-5p during MI/R, disclosed the specific mechanism of miR-17-5p/Tsg101 regulatory axis in ER stress-induced myocardium injury and cardiomyocytes apoptosis, and presented a promising diagnostic biomarker and potential target for therapy of AMI.
Collapse
Affiliation(s)
- Linlin Zhao
- Department of Cardiac Surgery, The People's Hospital of Liaoning Province, No.33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Shan Jiang
- Department of Respiration, Shengjing Hospital of China Medical University, Shenyang, 110000, Liaoning, People's Republic of China
| | - Naishi Wu
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Enyi Shi
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Lin Yang
- Department of Cardiovascular Medicine, The People's Hospital of Liaoning Province, Shenyang, 110016, Liaoning, People's Republic of China
| | - Qiang Li
- Department of Cardiac Surgery, The People's Hospital of Liaoning Province, No.33 Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
10
|
Sadek MS, Cachorro E, El-Armouche A, Kämmerer S. Therapeutic Implications for PDE2 and cGMP/cAMP Mediated Crosstalk in Cardiovascular Diseases. Int J Mol Sci 2020; 21:E7462. [PMID: 33050419 PMCID: PMC7590001 DOI: 10.3390/ijms21207462] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Phosphodiesterases (PDEs) are the principal superfamily of enzymes responsible for degrading the secondary messengers 3',5'-cyclic nucleotides cAMP and cGMP. Their refined subcellular localization and substrate specificity contribute to finely regulate cAMP/cGMP gradients in various cellular microdomains. Redistribution of multiple signal compartmentalization components is often perceived under pathological conditions. Thereby PDEs have long been pursued as therapeutic targets in diverse disease conditions including neurological, metabolic, cancer and autoimmune disorders in addition to numerous cardiovascular diseases (CVDs). PDE2 is a unique member of the broad family of PDEs. In addition to its capability to hydrolyze both cAMP and cGMP, PDE2 is the sole isoform that may be allosterically activated by cGMP increasing its cAMP hydrolyzing activity. Within the cardiovascular system, PDE2 serves as an integral regulator for the crosstalk between cAMP/cGMP pathways and thereby may couple chronically adverse augmented cAMP signaling with cardioprotective cGMP signaling. This review provides a comprehensive overview of PDE2 regulatory functions in multiple cellular components within the cardiovascular system and also within various subcellular microdomains. Implications for PDE2- mediated crosstalk mechanisms in diverse cardiovascular pathologies are discussed highlighting the prospective use of PDE2 as a potential therapeutic target in cardiovascular disorders.
Collapse
Affiliation(s)
| | | | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| | - Susanne Kämmerer
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| |
Collapse
|
11
|
CaMKIIδ inhibition protects against myocardial ischemia/reperfusion injury: Role of Beclin-1-dependent autophagy. Eur J Pharmacol 2020; 886:173539. [PMID: 32918874 DOI: 10.1016/j.ejphar.2020.173539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 01/11/2023]
Abstract
Ca2+/calmodulin-dependent protein kinase II δ (CaMKIIδ) has been shown to play a vital role in pathological events in myocardial ischemia/reperfusion (IR) injury. Dysregulation of autophagy in cardiomyocytes is implicated in myocardial IR injury. Here, we examined whether CaMKIIδ inhibition could protect against myocardial IR injury through alleviating autophagy dysfunction and evaluated the potential role of CaMKIIδ in Beclin-1-dependent autophagy in ischemia/reperfused hearts. This study was performed using isolated perfused rat hearts and H9c2 cardiac myoblasts. KN-93, but not KN-92, inhibited the phosphorylation of CaMKIIδ at Thr286 and its substrate phospholamban at Thr17 besides the CaMKIIδ activity in myocardial IR. KN-93, but not KN-92 significantly improved post-ischemic cardiac function and reduced cell death. In cultured H9c2 cardiac myoblasts, KN-93 or CaMKIIδ siRNA, but not KN-92, attenuated simulated IR (SIR)-induced cell death. Moreover, CaMKIIδ inhibition could alleviate IR-induced autophagic dysfunction as evidenced in reduced levels of Atg5, p62, and LC3BII in isolated rat hearts and H9c2 cardiac myoblasts. Furthermore, co-treatment with bafilomycin A1, a lysosomal inhibitor, in CaMKII inhibition-treated cells suggested that CaMKII inhibition alleviated autophagic flux. CaMKIIδ inhibition mitigated the phosphorylation of Beclin-1 at Ser90. As expected, Beclin-1 siRNA significantly decreased the levels of Beclin-1 and Beclin-1 phosphorylation accompanied by partial reductions in Atg5, LC3BII, p62, cleaved caspase-3 and cytochrome c. However, Beclin-1 siRNA had little effect on CaMKIIδ phosphorylation. Taken together, these results demonstrated that CaMKIIδ inhibition reduced myocardial IR injury by improving autophagy dysfunction, and that CaMKIIδ-induced autophagy dysfunction partially depended on the phosphorylation of Beclin-1.
Collapse
|
12
|
Meng Z, Gai W, Song D. Postconditioning with Nitrates Protects Against Myocardial Reperfusion Injury: A New Use for an Old Pharmacological Agent. Med Sci Monit 2020; 26:e923129. [PMID: 32516304 PMCID: PMC7299064 DOI: 10.12659/msm.923129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Early reperfusion remains the key therapy to salvage viable myocardium and must be applied as soon as possible following an acute myocardial infarction (AMI) to attenuate the ischemic insult. However, reperfusion injury may develop following reintroduction of blood and oxygen to vulnerable myocytes, which results in more severe cell death than in the preceding ischemic episode. Ischemic postconditioning (I-PostC) provides a cardioprotective effect in combination with pharmacological agents. Although nitrates have been tested in many experimental and clinical studies of acute AMI to evaluate the cardioprotective effect, few investigations have been focused on nitrates postconditioning in patients undergoing percutaneous coronary intervention (PCI). This review presents the manifestations of myocardial reperfusion injury (RI) and potential mechanisms underlying it, and provides the mechanisms involved in the cardioprotection of I-PostC. We also present a new therapeutic approach to attenuate RI by use of an ‘old’ agent – nitrates – in AMI patients.
Collapse
Affiliation(s)
- Zhu Meng
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| | - Weili Gai
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| | - Dalin Song
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| |
Collapse
|
13
|
Davidson SM, Adameová A, Barile L, Cabrera-Fuentes HA, Lazou A, Pagliaro P, Stensløkken KO, Garcia-Dorado D. Mitochondrial and mitochondrial-independent pathways of myocardial cell death during ischaemia and reperfusion injury. J Cell Mol Med 2020; 24:3795-3806. [PMID: 32155321 PMCID: PMC7171390 DOI: 10.1111/jcmm.15127] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/08/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myocardial infarction causes lethal injury to cardiomyocytes during both ischaemia and reperfusion (IR). It is important to define the precise mechanisms by which they die in order to develop strategies to protect the heart from IR injury. Necrosis is known to play a major role in myocardial IR injury. There is also evidence for significant myocardial death by other pathways such as apoptosis, although this has been challenged. Mitochondria play a central role in both of these pathways of cell death, as either a causal mechanism is the case of mitochondrial permeability transition leading to necrosis, or as part of the signalling pathway in mitochondrial cytochrome c release and apoptosis. Autophagy may impact this process by removing dysfunctional proteins or even entire mitochondria through a process called mitophagy. More recently, roles for other programmed mechanisms of cell death such as necroptosis and pyroptosis have been described, and inhibitors of these pathways have been shown to be cardioprotective. In this review, we discuss both mitochondrial and mitochondrial‐independent pathways of the major modes of cell death, their role in IR injury and their potential to be targeted as part of a cardioprotective strategy. This article is part of a special Issue entitled ‘Mitochondria as targets of acute cardioprotection’ and emerged as part of the discussions of the European Union (EU)‐CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Adriana Adameová
- Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia.,Centre of Experimental Medicine SAS, Bratislava, Slovakia
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation and Faculty of Biomedical Sciences, Università Svizzera Italiana, Lugano, Switzerland
| | - Hector Alejandro Cabrera-Fuentes
- SingHealth Duke-NUS Cardiovascular Sciences Academic Clinical Programme and Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, Nuevo Leon, México.,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia.,Institute of Physiology, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pasquale Pagliaro
- Department of Biological and Clinical Sciences, University of Turin, Torino, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| | - Kåre-Olav Stensløkken
- Section of Physiology, Department of Molecular Medicine, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - David Garcia-Dorado
- IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain.,Department of Cardiology, Vascular Biology and Metabolism Area, Vall d'Hebron University Hospital and Research Institute (VHIR), Barcelona, Spain.,Universitat Autónoma de Barcelona, Barcelona, Spain
| | | |
Collapse
|
14
|
Qin QJ, Cui LQ, Li P, Wang YB, Zhang XZ, Guo ML. Rhynchophylline ameliorates myocardial ischemia/reperfusion injury through the modulation of mitochondrial mechanisms to mediate myocardial apoptosis. Mol Med Rep 2019; 19:2581-2590. [PMID: 30720139 PMCID: PMC6423601 DOI: 10.3892/mmr.2019.9908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 01/18/2019] [Indexed: 01/22/2023] Open
Abstract
Rhynchophylline (RP), the primary active ingredient of Uncaria rhynchophylla, has an anti-hypertensive effect and protects against ischemia-induced neuronal damage. The present study aimed to examine the roles and mechanisms of RP in myocardial ischemia-reperfusion (MI/R) injury of rat cardiomyocytes. Cell viability, reactive oxygen species, mitochondrial membrane potential (MMP) and cell apoptosis were examined by a Cell Counting Kit-8 assay and flow cytometry, respectively. An ELISA was performed to assess the expression of oxidative stress markers. Spectrophotometry was used to detect the degree of mitochondrial permeability transition pore (mPTP) openness. Western blotting and reverse transcription- quantitative polymerase chain reaction assays were used to evaluate the associated protein and mRNA expression, respectively. The present results demonstrated that RP increased the cell viability of MI/R-induced cardiomyocytes, and suppressed the MI/R-induced apoptosis of cardiomyocytes. Additionally, RP modulated the Ca2+ and MMP levels in MI/R-induced cardiomyocytes. Furthermore, RP decreased the oxidative stress and mPTP level of MI/R-induced cardiomyocytes. It was additionally observed that RP affected the apoptosis-associated protein expression and regulated the mitochondrial-associated gene expression in MI/R-induced cardiomyocytes. In conclusion, RP ameliorated MI/R injury through the modulation of mitochondrial mechanisms. The potential effects of RP on the protection of MI/R-induced apoptosis of cardiomyocytes suggest that RP may be an effective target for MI/R therapy.
Collapse
Affiliation(s)
- Qiao-Ji Qin
- Emergency Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Li-Qiang Cui
- Department of Anesthesiology, Chengyang People's Hospital, Qingdao, Shandong 266109, P.R. China
| | - Peng Li
- Emergency Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yong-Bin Wang
- Emergency Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xue-Zhi Zhang
- Emergency Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ming-Lei Guo
- Emergency Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
15
|
Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Mitochondrial bioenergetics and cardiolipin alterations in myocardial ischemia-reperfusion injury: implications for pharmacological cardioprotection. Am J Physiol Heart Circ Physiol 2018; 315:H1341-H1352. [PMID: 30095969 DOI: 10.1152/ajpheart.00028.2018] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mitochondrial dysfunction plays a central role in myocardial ischemia-reperfusion (I/R) injury. Increased reactive oxygen species production, impaired electron transport chain activity, aberrant mitochondrial dynamics, Ca2+ overload, and opening of the mitochondrial permeability transition pore have been proposed as major contributory factors to mitochondrial dysfunction during myocardial I/R injury. Cardiolipin (CL), a mitochondria-specific phospholipid, plays a pivotal role in multiple mitochondrial bioenergetic processes, including respiration and energy conversion, in mitochondrial morphology and dynamics as well as in several steps of the apoptotic process. Changes in CL levels, species composition, and degree of oxidation may have deleterious consequences for mitochondrial function with important implications in a variety of pathophysiological conditions, including myocardial I/R injury. In this review, we focus on the role played by CL alterations in mitochondrial dysfunction in myocardial I/R injury. Pharmacological strategies to prevent myocardial injury during I/R targeting mitochondrial CL are also examined.
Collapse
Affiliation(s)
- Giuseppe Paradies
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari , Bari , Italy
| | | | - Francesca Maria Ruggiero
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari , Bari , Italy
| | - Giuseppe Petrosillo
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, National Research Council , Bari , Italy
| |
Collapse
|
16
|
Rinaldi L, Pozdniakova S, Jayarajan V, Troidl C, Abdallah Y, Aslam M, Ladilov Y. Protective role of soluble adenylyl cyclase against reperfusion-induced injury of cardiac cells. Biochim Biophys Acta Mol Basis Dis 2018; 1865:252-260. [PMID: 30044950 DOI: 10.1016/j.bbadis.2018.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/15/2018] [Accepted: 07/17/2018] [Indexed: 12/16/2022]
Abstract
AIMS Disturbance of mitochondrial function significantly contributes to the myocardial injury that occurs during reperfusion. Increasing evidence suggests a role of intra-mitochondrial cyclic AMP (cAMP) signaling in promoting respiration and ATP synthesis. Mitochondrial levels of cAMP are controlled by type 10 soluble adenylyl cyclase (sAC) and phosphodiesterase 2 (PDE2), however their role in the reperfusion-induced injury remains unknown. Here we aimed to examine whether sAC may support cardiomyocyte survival during reperfusion. METHODS AND RESULTS Adult rat cardiomyocytes or rat cardiac H9C2 cells were subjected to metabolic inhibition and recovery as a model of simulated ischemia and reperfusion. Cytosolic Ca2+, pH, mitochondrial cAMP (live-cell imaging), and cell viability were analyzed during a 15-min period of reperfusion. Suppression of sAC activity in cardiomyocytes and H9C2 cells, either by sAC knockdown, by pharmacological inhibition or by withdrawal of bicarbonate, a natural sAC activator, compromised cell viability and recovery of cytosolic Ca2+ homeostasis during reperfusion. Contrariwise, overexpression of mitochondria-targeted sAC in H9C2 cells suppressed reperfusion-induced cell death. Analyzing cAMP concentration in mitochondrial matrix we found that inhibition of PDE2, a predominant mitochondria-localized PDE isoform in mammals, during reperfusion significantly increased cAMP level in mitochondrial matrix, but not in cytosol. Accordingly, PDE2 inhibition attenuated reperfusion-induced cardiomyocyte death and improved recovery of the cytosolic Ca2+ homeostasis. CONCLUSION sAC plays an essential role in supporting cardiomyocytes viability during reperfusion. Elevation of mitochondrial cAMP pool either by sAC overexpression or by PDE2 inhibition beneficially affects cardiomyocyte survival during reperfusion.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
| | | | | | - Christian Troidl
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
| | - Yaser Abdallah
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
| | - Muhammad Aslam
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany; DZHK (German Centre for Cardiovascular Research), partner site Rhein-Main, Bad Nauheim, Germany
| | - Yury Ladilov
- Center for Cardiovascular Research, Charité, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
17
|
Tanaka H, Okuda K, Ohtani S, Asari M, Horioka K, Isozaki S, Hayakawa A, Ogawa K, Hiroshi S, Shimizu K. Chained nuclei and python pattern in skeletal muscle cells as histological markers for electrical injury. Leg Med (Tokyo) 2017; 32:75-78. [PMID: 29579533 DOI: 10.1016/j.legalmed.2017.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/26/2017] [Accepted: 11/01/2017] [Indexed: 10/18/2022]
Abstract
Electrical injury is damage caused by an electrical current passing through the body. We have previously reported that irregular stripes crossing skeletal muscle fibers (python pattern) and multiple small nuclei arranged in the longitudinal direction of the muscle fibers (chained nuclear change) are uniquely observed by histopathological analysis in the skeletal muscle tissues of patients with electrical injury. However, it remains unclear whether these phenomena are caused by the electrical current itself or by the joule heat generated by the electric current passing through the body. To clarify the causes underlying these changes, we applied electric and heat injury to the exteriorized rat soleus muscle in situ. Although both the python pattern and chained nuclear change were induced by electric injury, only the python pattern was induced by heat injury. Furthermore, a chained nuclear change was induced in the soleus muscle cells by electric current flow in physiological saline at 40 °C ex vivo, but a python pattern was not observed. When the skeletal muscle was exposed to electrical injury in cardiac-arrested rats, a python pattern was induced within 5 h after cardiac arrest, but no chained nuclear change was observed. Therefore, a chained nuclear change is induced by an electrical current alone in tissues in vital condition, whereas a python pattern is caused by joule heat, which may occur shortly after death. The degree and distribution of these skeletal muscle changes may be useful histological markers for analyzing cases of electrical injury in forensic medicine.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Department of Legal Medicine, Asahikawa Medical University, Japan
| | - Katsuhiro Okuda
- Department of Legal Medicine, Asahikawa Medical University, Japan
| | - Seiji Ohtani
- Department of Legal Medicine, Asahikawa Medical University, Japan.
| | - Masaru Asari
- Department of Legal Medicine, Asahikawa Medical University, Japan
| | - Kie Horioka
- Department of Legal Medicine, Asahikawa Medical University, Japan
| | | | - Akira Hayakawa
- Tokyo Medical Examiner's Office, Tokyo Metropolitan Government, Japan
| | - Katsuhiro Ogawa
- Department of Pathology, Asahikawa Medical University, Japan
| | - Shiono Hiroshi
- Department of Legal Medicine, Asahikawa Medical University, Japan
| | - Keiko Shimizu
- Department of Legal Medicine, Asahikawa Medical University, Japan
| |
Collapse
|
18
|
Li J, Yan Z, Fang Q. A Mechanism Study Underlying the Protective Effects of Cyclosporine-A on Lung Ischemia-Reperfusion Injury. Pharmacology 2017; 100:83-90. [PMID: 28501872 DOI: 10.1159/000458760] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/03/2017] [Indexed: 11/19/2022]
Abstract
AIM This study is aimed at validating the hypothesis that administration of cyclosporine-A (CsA) would be protective in lung ischemia-reperfusion (I/R) injury and in exploring the underlying mechanism. METHODS Rabbits were divided into 4 groups: the control, sham operation, I/R, and I/R with CsA treatment. Flow cytometry was used to measure the mitochondrial membrane potential. Laser scanning confocal microscope was used to analyze mitochondrion permeability transition pore (MPTP). The apoptotic cell was detected by the TUNEL staining. Western blot was performed to analyze the protein expression levels. RESULTS CsA not only attenuated the histopathologic alterations in lung and mitochondria after I/R injury, but also attenuated I/R injury through increasing MPP and inhibiting MPTP opening. Besides, CsA attenuated I/R injury through suppressing the release of cytochrome-c (CytC), inhibiting cell apoptosis and decreasing the expression levels of cyclophilin-D (Cyp-D), adenine nucleotide translocase 1 (ANT1) and voltage-dependent anion channel 1 (VDAC1). Finally, we found that Cyp-D knockdown inhibits I/R injury-induced MPTP opening and cell apoptosis. CONCLUSION Our study found that the protective role of CsA on lung I/R injury depends on the inhibition of MPTP and CytC release, suppression of the activation of mitochondrial apoptosis pathway and the expressions of apoptotic-related proteins, as well as the decreased expression levels of ANT1 and VDAC1.
Collapse
Affiliation(s)
- Jian''an Li
- Department of Cardiac Surgery, Anhui Provincial Hospital, Anhui Medical University, Anhui, PR China
| | | | | |
Collapse
|
19
|
Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Cardiolipin alterations and mitochondrial dysfunction in heart ischemia/reperfusion injury. ACTA ACUST UNITED AC 2015. [DOI: 10.2217/clp.15.31] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Alburquerque-Béjar JJ, Barba I, Inserte J, Miró-Casas E, Ruiz-Meana M, Poncelas M, Vilardosa Ú, Valls-Lacalle L, Rodríguez-Sinovas A, Garcia-Dorado D. Combination therapy with remote ischaemic conditioning and insulin or exenatide enhances infarct size limitation in pigs. Cardiovasc Res 2015; 107:246-54. [PMID: 26045476 DOI: 10.1093/cvr/cvv171] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/23/2015] [Indexed: 12/18/2022] Open
Abstract
AIMS Remote ischaemic conditioning (RIC) has been shown to reduce myocardial infarct size in patients. Our objective was to investigate whether the combination of RIC with either exenatide or glucose-insulin-potassium (GIK) is more effective than RIC alone. METHODS AND RESULTS Pigs were submitted to 40 min of coronary occlusion followed by reperfusion, and received (i) no treatment, (ii) one of the following treatments: RIC (5 min ischemia/5 min reperfusion × 4), GIK, or exenatide (at doses reducing infarct size in clinical trials), or (iii) a combination of two of these treatments (RIC + GIK or RIC + exenatide). After 5 min of reperfusion (n = 4/group), prominent phosphorylation of Akt and endothelial nitric oxide synthase (eNOS) was observed, both in control and reperfused myocardium, in animals receiving GIK, and mitochondria from these hearts showed reduced ADP-stimulated respiration. (1)H NMR-based metabonomics disclosed a shift towards increased glycolysis in GIK and exenatide groups. In contrast, oxidative stress (myocardial nitrotyrosine levels) and eNOS uncoupling were significantly reduced only by RIC. In additional experiments (n = 7-10/group), ANOVA demonstrated a significant effect of the number of treatments after 2 h of reperfusion on infarct size (triphenyltetrazolium, % of the area at risk; 59.21 ± 3.34, 36.64 ± 3.03, and 21.04 ± 2.38% for none, one, and two treatments, respectively), and significant differences between one and two treatments (P = 0.004) but not among individual treatments or between RIC + GIK and RIC + exenatide. CONCLUSIONS GIK and exenatide activate cardioprotective pathways different from those of RIC, and have additive effects with RIC on infarct size reduction in pigs.
Collapse
Affiliation(s)
- Juan José Alburquerque-Béjar
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ignasi Barba
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Javier Inserte
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Elisabet Miró-Casas
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marisol Ruiz-Meana
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marcos Poncelas
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Úrsula Vilardosa
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Laura Valls-Lacalle
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - David Garcia-Dorado
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
21
|
Aoyagi T, Higa JK, Aoyagi H, Yorichika N, Shimada BK, Matsui T. Cardiac mTOR rescues the detrimental effects of diet-induced obesity in the heart after ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2015; 308:H1530-9. [PMID: 25888508 DOI: 10.1152/ajpheart.00008.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/10/2015] [Indexed: 01/22/2023]
Abstract
Diet-induced obesity deteriorates the recovery of cardiac function after ischemia-reperfusion (I/R) injury. While mechanistic target of rapamycin (mTOR) is a key mediator of energy metabolism, the effects of cardiac mTOR in ischemic injury under metabolic syndrome remains undefined. Using cardiac-specific transgenic mice overexpressing mTOR (mTOR-Tg mice), we studied the effect of mTOR on cardiac function in both ex vivo and in vivo models of I/R injury in high-fat diet (HFD)-induced obese mice. mTOR-Tg and wild-type (WT) mice were fed a HFD (60% fat by calories) for 12 wk. Glucose intolerance and insulin resistance induced by the HFD were comparable between WT HFD-fed and mTOR-Tg HFD-fed mice. Functional recovery after I/R in the ex vivo Langendorff perfusion model was significantly lower in HFD-fed mice than normal chow diet-fed mice. mTOR-Tg mice demonstrated better cardiac function recovery and had less of the necrotic markers creatine kinase and lactate dehydrogenase in both feeding conditions. Additionally, mTOR overexpression suppressed expression of proinflammatory cytokines, including IL-6 and TNF-α, in both feeding conditions after I/R injury. In vivo I/R models showed that at 1 wk after I/R, HFD-fed mice exhibited worse cardiac function and larger myocardial scarring along myofibers compared with normal chow diet-fed mice. In both feeding conditions, mTOR overexpression preserved cardiac function and prevented myocardial scarring. These findings suggest that cardiac mTOR overexpression is sufficient to prevent the detrimental effects of diet-induced obesity on the heart after I/R, by reducing cardiac dysfunction and myocardial scarring.
Collapse
Affiliation(s)
- Toshinori Aoyagi
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Jason K Higa
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Hiroko Aoyagi
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Naaiko Yorichika
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Briana K Shimada
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Takashi Matsui
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|
22
|
Abstract
Pathological contraction bands (CBs) are a type of necrosis pattern found in the myocardium. The composition of CB is not well studied. This is because CBs have diverse forms and can be observed in various causes of death. In pathology fields, CBs are classified artifactual CB and CB necrosis. We have identified different forms of CB by examining the expression and distribution of complement component C9 (CCC9) and Sirtuin1 by immunohistochemistry in the myocardium of patients who died because of different causes of death. We used cardiac tissues with CB from 30 forensic autopsy cases in our department from the last 2 years. We excluded the cases that had very little CB. We found that our CB classification based on expression levels of both CCC9 and Sirtuin1 correlated well with the agonal situation, including high temperature, myocardial infarction, cardiopulmonary resuscitation, and hypothermia. On the basis of these results, we here advocate a classification scheme based on immunohistochemistry. Furthermore, we found that CB necrosis could be detected using immunostaining with CCC9. Using our classification scheme, it will be possible to more accurately research each type of CB and the causative mechanisms.
Collapse
|
23
|
Helle KB, Corti A. Chromogranin A: a paradoxical player in angiogenesis and vascular biology. Cell Mol Life Sci 2015; 72:339-48. [PMID: 25297920 PMCID: PMC11113878 DOI: 10.1007/s00018-014-1750-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/10/2014] [Accepted: 09/29/2014] [Indexed: 12/18/2022]
Abstract
Half a century after the discovery of chromogranin A as a secreted product of the catecholamine storage granules in the bovine adrenal medulla, the physiological role for the circulating pool of this protein has been recently coined, namely as an important player in vascular homeostasis. While the circulating chromogranin A since 1984 has proved to be a significant and useful marker of a wide range of pathophysiological and pathological conditions involving the diffuse neuroendocrine system, this protein has now been assigned a physiological "raison d'etre" as a regulator in vascular homeostasis. Moreover, chromogranin A processing in response to tissue damage and blood coagulation provides the first indication of a difference in time frame of the regulation of angiogenesis evoked by the intact chromogranin A and its two major peptide products, vasostatin-1 and catestatin. The impact of these discoveries on vascular homeostasis, angiogenesis, cancer, tissue repair and cardio-regulation will be discussed.
Collapse
Affiliation(s)
- Karen B. Helle
- Department of Biomedicine, University of Bergen, Haukelandsvei 1, 5009 Bergen, Norway
| | - Angelo Corti
- Division of Oncology, San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| |
Collapse
|
24
|
Kim DE, Kim B, Shin HS, Kwon HJ, Park ES. The protective effect of hispidin against hydrogen peroxide-induced apoptosis in H9c2 cardiomyoblast cells through Akt/GSK-3β and ERK1/2 signaling pathway. Exp Cell Res 2014; 327:264-75. [PMID: 25128810 DOI: 10.1016/j.yexcr.2014.07.037] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 10/24/2022]
Abstract
Hispidin, a phenolic compound from Phellinus linteus (a medicinal mushroom), has been shown to possess strong anti-oxidant, anti-cancer, anti-diabetic, and anti-dementia properties. However, the cardioprotective efficacy of hispidin has not yet been investigated. In the present study, we investigated the protective effect of hispidin against oxidative stress-induced apoptosis in H9c2 cardiomyoblast cells and neonatal rat ventricular myocytes. While the treatment of H9c2 cardiomyoblast cells with hydrogen peroxide caused a loss of cell viability and an increase in the number of apoptotic cells, hispidin significantly protected the cells against hydrogen peroxide-induced cell death without any cytotoxicity as determined by XTT assay, LDH release assay, Hoechst 33342 assay, and Western blotting of apoptosis proteins such as caspase-3, Bax, and Bcl-2. Our data also shows that hispidin significantly scavenged intracellular ROS, and markedly enhanced the expression of antioxidant enzymes such as heme oxygenase-1 and catalase, which was accompanied by the concomitant activation of Akt/GSK-3β and ERK1/2 phosphorylation in H9c2 cardiomyoblast cells. The effects of hispidin on Akt and ERK phosphorylation were abrogated by LY294002 (a PI3K/Akt inhibitor) and U0126 (an ERK1/2 inhibitor). The effect of hispidin on GSK-3b activities was also blocked by LY294002. Furthermore, inhibiting the Akt/GSK-3β and ERK1/2 pathway by these inhibitors significantly reversed the hispidin-induced Bax and Bcl-2 expression, apoptosis induction, and ROS production. These findings indicate that hispidin protects against apoptosis in H9c2 cardiomyoblast cells exposed to hydrogen peroxide through reducing intracellular ROS production, regulating apoptosis-related proteins, and the activation of the Akt/GSK-3β and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Dae-Eun Kim
- Department of Biomaterials Science and Engineering, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Biomedical Laboratory Science, Kyungbok University, Sinbuk-myeon, Pochen, Gyeonggi 487-717, Republic of Korea
| | - Bokyung Kim
- Department of Physiology, Institute of Functional Genomics, Konkuk University School of Medicine, Chungju, Chungbuk 380-701, Republic of Korea
| | - Hwa-Sup Shin
- Department of Biomedical Chemistry, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Ho Jeong Kwon
- Department of Biomaterials Science and Engineering, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea.
| | - Eun-Seok Park
- Department of Biomedical Laboratory Science, Kyungbok University, Sinbuk-myeon, Pochen, Gyeonggi 487-717, Republic of Korea.
| |
Collapse
|
25
|
Morita S, Furukawa S, Nishi K. Immunohistochemical evaluation of hypoxia markers in the myocardium. AUST J FORENSIC SCI 2014. [DOI: 10.1080/00450618.2014.906653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
26
|
Garcia-Dorado D, Rodríguez-Sinovas A, Ruiz-Meana M, Inserte J. Protección contra el daño miocárdico por isquemia-reperfusión en la práctica clínica. Rev Esp Cardiol 2014. [DOI: 10.1016/j.recesp.2014.01.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
27
|
Garcia-Dorado D, Rodríguez-Sinovas A, Ruiz-Meana M, Inserte J. Protection against myocardial ischemia-reperfusion injury in clinical practice. ACTA ACUST UNITED AC 2014; 67:394-404. [PMID: 24774733 DOI: 10.1016/j.rec.2014.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 01/22/2014] [Indexed: 12/28/2022]
Abstract
Even when reperfusion therapy is applied as early as possible, survival and quality of life are compromised in a considerable number of patients with ST-segment elevation acute myocardial infarction. Some cell death following transient coronary occlusion occurs during reperfusion, due to poor handling of calcium in the sarcoplasmic reticulum-mitochondria system, calpain activation, oxidative stress, and mitochondrial failure, all promoted by rapid normalization of intracellular pH. Various clinical trials have shown that infarct size can be limited by nonpharmacological strategies--such as ischemic postconditioning and remote ischemic conditioning--or by drugs--such as cyclosporine, insulin, glucagon-like peptide-1 agonists, beta-blockers, or stimulation of cyclic guanosine monophosphate synthesis. However, some clinical studies have yielded negative results, largely due to a lack of consistent preclinical data or a poor design, especially delayed administration. Large-scale clinical trials are therefore necessary, particularly those with primary clinical variables and combined therapies that consider age, sex, and comorbidities, to convert protection against reperfusion injury into a standard treatment for patients with ST-segment elevation acute myocardial infarction.
Collapse
Affiliation(s)
- David Garcia-Dorado
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Antonio Rodríguez-Sinovas
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marisol Ruiz-Meana
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Javier Inserte
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
28
|
Huang K, Lu SJ, Zhong JH, Xiang Q, Wang L, Wu M. Comparative analysis of different cyclosporine A doses on protection after myocardial ischemia/reperfusion injury in rat. ASIAN PAC J TROP MED 2014; 7:144-8. [DOI: 10.1016/s1995-7645(14)60011-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/15/2013] [Accepted: 01/15/2014] [Indexed: 10/25/2022] Open
|
29
|
Lindholm MG, Boesgaard S, Thune JJ, Kelbaek H, Andersen HR, Kober L. Percutaneous coronary intervention for acute MI does not prevent in-hospital development of cardiogenic shock compared to fibrinolysis. Eur J Heart Fail 2014; 10:668-74. [DOI: 10.1016/j.ejheart.2008.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 04/08/2008] [Accepted: 04/28/2008] [Indexed: 11/28/2022] Open
Affiliation(s)
- Matias G. Lindholm
- Medical Department B, Division of Cardiology; Rigshospitalet, University Hospital of Copenhagen; Denmark
| | - Søren Boesgaard
- Medical Department B, Division of Cardiology; Rigshospitalet, University Hospital of Copenhagen; Denmark
| | - Jens Jakob Thune
- Medical Department B, Division of Cardiology; Rigshospitalet, University Hospital of Copenhagen; Denmark
| | - Henning Kelbaek
- Medical Department B, Division of Cardiology; Rigshospitalet, University Hospital of Copenhagen; Denmark
| | - Henning Rud Andersen
- Department of Cardiology, Skejby Hospital; University Hospital of Aarhus; Denmark
| | - Lars Kober
- Medical Department B, Division of Cardiology; Rigshospitalet, University Hospital of Copenhagen; Denmark
| | | |
Collapse
|
30
|
Lim KH, Lim DJ, Kim JH. Ginsenoside-Re ameliorates ischemia and reperfusion injury in the heart: a hemodynamics approach. J Ginseng Res 2013; 37:283-92. [PMID: 24198653 PMCID: PMC3818954 DOI: 10.5142/jgr.2013.37.283] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/18/2013] [Accepted: 02/18/2013] [Indexed: 11/18/2022] Open
Abstract
Ginsenosides are divided into two groups based on the types of the panaxadiol group (e.g., ginsenoside-Rb1 and -Rc) and the panaxatriol group (e.g., ginsenoside-Rg1 and -Re). Among them, ginsenoside-Re (G-Re) is one of the compounds with the highest content in Panax ginseng and is responsible for pharmacological effects. However, it is not yet well reported if G-Re increases the hemodynamics functions on ischemia (30 min)/reperfusion (120 min) (I/R) induction. Therefore, in the present study, we investigated whether treatment of G-Re facilitated the recovery of hemodynamic parameters (heart rate, perfusion pressure, aortic flow, coronary flow, and cardiac output) and left ventricular developed pressure (±dp/dtmax). This research is designed to study the effects of G-Re by studying electrocardiographic changes such as QRS interval, QT interval and R-R interval, and inflammatory marker such as tissue necrosis factor-α (TNF-α) in heart tissue in I/R-induced heart. From the results, I/R induction gave a significant increase in QRS interval, QT interval and R-R interval, but showed decrease in all hemodynamic parameters. I/R induction resulted in increased TNF-α level. Treatment of G-Re at 30 and 100 μM doses before I/R induction significantly prevented the decrease in hemodynamic parameters, ameliorated the electrocardiographic abnormality, and inhibited TNF-α level. In this study, G-Re at 100 μM dose exerted more beneficial effects on cardiac function and preservation of myocardium in I/R injury than 30 μM. Collectively, these results indicate that G-Re has distinct cardioprotectective effects in I/R induced rat heart.
Collapse
Affiliation(s)
- Kyu Hee Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Jeonju 561-756, Korea
| | | | | |
Collapse
|
31
|
Yeh CH, Chen TP, Wang YC, Fang SW, Wun TC. Potent cardioprotection from ischemia-reperfusion injury by a two-domain fusion protein comprising annexin V and Kunitz protease inhibitor. J Thromb Haemost 2013; 11:1454-63. [PMID: 23746209 PMCID: PMC3752160 DOI: 10.1111/jth.12314] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 05/25/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND Considerable evidence suggests that coagulation proteases (tissue factor [TF]/activated factor VII [FVIIa]/FXa/thrombin) and their target protease activated receptors (PAR-1/PAR-2) play important roles in myocardial ischemia-reperfusion (I-R) injury. We hypothesized that localized inhibition of TF/FVIIa on the membrane surfaces of ischemic cells could effectively block coagulation cascade and subsequent PAR-1/PAR-2 cell signaling, thereby protecting the myocardium from I-R injury. OBJECTIVES We recently developed an annexin V-Kunitz inhibitor fusion protein (ANV-6L15) that could specifically bind to anionic phospholipids on the membrane surfaces of apoptotic cells and efficiently inhibit the membrane-anchored TF/FVIIa. In this study, we investigated the cardioprotective effect of ANV-6L15 in a rat cardiac I-R model in comparison with that of hirudin. METHODS Left coronary artery occlusion was maintained for 45 min followed by 4 h of reperfusion in anesthetized Sprague-Dawley rats. One minute before or 2 min after coronary ligation, rats received an intravenous bolus injection of ANV-6L15 (2.5-250 μg kg(-1) ), vehicle, or hirudin via bolus injection and continuous infusion. RESULTS AND CONCLUSIONS ANV-6L15 dose-dependently reduced infarct size by up to 87% and decreased plasma levels of cardiac troponin I, tumor necrosis factor-α, and soluble intercellular adhesion molecule-1, by up to 97%, 96%, and 66%, respectively, with little impact on the coagulation parameters. ANV-6L15 also ameliorated hemodynamic derangements, attenuated neutrophil infiltration and reduced Terminal deoxynucleotidyl transferase dUTP nick end labeling-positive apoptotic cardiomyocytes. Hirudin was less efficacious even at supraclinical dose. ANV-6L15 confers exceptionally potent cardioprotection and is a promising drug candidate for the prevention of myocardial I-R injury.
Collapse
Affiliation(s)
- Chi-Hsiao Yeh
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
- College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Tao-Yuan, Taiwan ROC 333
| | - Tzu-Ping Chen
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
| | - Yao-Chang Wang
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
| | - Shu-Wen Fang
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
| | - Tze-Chein Wun
- EVAS Therapeutics, LLC, 613 Huntley Heights Drive, Ballwin MO 63021, USA
| |
Collapse
|
32
|
Ravingerová T, Čarnická S, Nemčeková M, Ledvényiová V, Adameová A, Khandelwal VK, Zálešák M, Kolář F. The impact of lifestyle-related risk factors on cardiac response to ischemia and possibilities to restore impaired ischemic tolerance. Physiol Res 2013; 61:S1-10. [PMID: 23130893 DOI: 10.33549/physiolres.932396] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Risk factors (RF) of cardiovascular diseases associated with modern lifestyle, such as stress, chronically increased blood pressure, hyperglycemia and dyslipidemia have a negative impact on the heart exposed to ischemia: they may facilitate its lethal injury (myocardial infarction) and occurrence of sudden death due to ventricular arrhythmias. On the other hand, some stressful stimuli related to RF including reactive oxygen species, transient episodes of ischemia (hypoxia), high glucose and other may play a dual role in the pathogenesis of ischemia/reperfusion (I/R) injury (IRI). Besides their deleterious effects, these factors may trigger adaptive processes in the heart resulting in greater resistance against IRI, which is also a characteristic feature of the female myocardium. However, sensitivity to ischemia is increasing with age in both genders. Current research indicates that comorbidity related to lifestyle may impair the cardiac response to acute ischemia not only by interference with pathophysiological mechanisms of IRI per se, but via suppression of intrinsic protective mechanisms in the heart and its ability to tolerate the ischemic challenges, although the role of RF has not been unequivocally proven. Moreover, even pathologically altered myocardium need not completely lose its adaptive potential. In addition, increased ischemic tolerance can be induced by the pleiotropic (independent of the primary) effects of some hypolipidemic and antidiabetic drugs, even in the diseased myocardium. This review addresses the issue of the impact of RF on cellular cardioprotective mechanisms and the possibilities to restore adaptive potential in subjects challenged with several RF. Reactivation of adaptive processes in the myocardium taking into consideration gender and age can contribute to optimalization of antiischemic therapy.
Collapse
Affiliation(s)
- T Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence SAS NOREG, Bratislava, Slovakia.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
The effect of preoperative melatonin on nuclear erythroid 2-related factor 2 activation in patients undergoing coronary artery bypass grafting surgery. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:676829. [PMID: 23691266 PMCID: PMC3649755 DOI: 10.1155/2013/676829] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 03/03/2013] [Indexed: 12/21/2022]
Abstract
During and after coronary artery bypass grafting (CABG), oxidative stress occurs. Finding an effective way to improve antioxidant response is important in CABG surgery. It has been shown that patients with coronary heart disease have a low Melatonin production rate. The present study aimed to investigate the effects of Melatoninon nuclear erythroid 2-related factor 2(Nrf2) activity in patients undergoing CABG surgery. Thirty volunteers undergoing CABG were randomized to receive 10 mg oral Melatonin (Melatonin group, n = 15) or placebo (placebo group, n = 15) before sleeping for 1 month before surgery. The activated Nrf2 was measured twice by DNA-based ELISA method in the nuclear extract of peripheral blood mononuclear cells of patients before aortic clumps and 45 minutes after CABG operation. Melatonin administration was associated with a significant increase in both plasma levels of Melatonin and Nrf2 concentration in Melatonin group compared to placebo group, respectively (15.2 ± 4.6 pmol/L, 0.28 ± 0.01 versus 1.1 ± 0.59 pmol/L, 0.20 ± 0.07, P < 0.05). The findings of the present study provide preliminary data suggesting that Melatonin may play a significant role in the potentiation of the antioxidant defense and attenuate cellular damages resulting from CABG surgery via theNrf2 pathway.
Collapse
|
34
|
Abstract
The main determinant of myocardial necrosis following an acute myocardial infarction (AMI) is duration of ischemia. Infarct size is a strong independent predictor of postinfarction mortality. Interventions able to protect the myocardium from death during an AMI (cardioprotection) are urgently needed. Myocardial injury associated with reperfusion (ischemia/reperfusion injury [I/R]) significantly contributes to the final necrotic size. Duration of ischemia can only be reduced by social and emergency medical services--hospital collaborative programs. However, for a given duration of ischemia, infarct size can be limited by reducing reperfusion injury. Despite the fact that several therapies have been shown to reduce I/R injury in animal models, translation to humans has been frustrating. The cost of developing new drugs able to reduce I/R injury is huge, and this is a major roadblock in the field of cardioprotection. Recent studies have proposed that old, inexpensive drugs--in human use for decades (e.g., β-blockers and cyclosporine, among others)--can reduce I/R injury when administered intravenously before coronary opening. The demonstration of such a cardioprotective effect should have a significant impact in the care of AMI patients.
Collapse
Affiliation(s)
- David Sanz-Rosa
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | | |
Collapse
|
35
|
Prunier F, Bière L, Gilard M, Boschat J, Mouquet F, Bauchart JJ, Charbonnier B, Genée O, Guérin P, Warin-Fresse K, Durand E, Lafont A, Christiaens L, Abi-Khalil W, Delépine S, Benard T, Furber A. Single high-dose erythropoietin administration immediately after reperfusion in patients with ST-segment elevation myocardial infarction: results of the erythropoietin in myocardial infarction trial. Am Heart J 2012; 163:200-7.e1. [PMID: 22305837 DOI: 10.1016/j.ahj.2011.11.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 11/07/2011] [Indexed: 12/22/2022]
Abstract
BACKGROUND Preclinical studies and pilot clinical trials have shown that high-dose erythropoietin (EPO) reduces infarct size in acute myocardial infarction. We investigated whether a single high-dose of EPO administered immediately after reperfusion in patients with ST-segment elevation myocardial infarction (STEMI) would limit infarct size. METHODS A total of 110 patients undergoing successful primary coronary intervention for a first STEMI was randomized to receive standard care either alone (n = 57) or combined with intravenous administration of 1,000 U/kg of epoetin β immediately after reperfusion (n = 53). The primary end point was infarct size assessed by gadolinium-enhanced cardiac magnetic resonance after 3 months. Secondary end points included left ventricular (LV) volume and function at 5-day and 3-month follow-up, incidence of microvascular obstruction (MVO), and safety. RESULTS Erythropoietin significantly decreased the incidence of MVO (43.4% vs 65.3% in the control group, P = .03) and reduced LV volume, mass, and function impairment at 5-day follow-up (all P < .05). After 3 months, median infarct size (interquartile range) was 17.5 g (7.6-26.1 g) in the EPO group and 16.0 g (9.4-28.2 g) in the control group (P = .64); LV mass, volume, and function were not significantly different between the 2 groups. The same number of major adverse cardiac events occurred in both groups. CONCLUSIONS Single high-dose EPO administered immediately after successful reperfusion in patients with STEMI did not reduce infarct size at 3-month follow-up. However, this regimen decreased the incidence of MVO and was associated with transient favorable effects on LV volume and function.
Collapse
|
36
|
Miura T, Tanno M. The mPTP and its regulatory proteins: final common targets of signalling pathways for protection against necrosis. Cardiovasc Res 2011; 94:181-9. [PMID: 22072634 DOI: 10.1093/cvr/cvr302] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The mitochondrial permeability transition pore (mPTP) is a non-selective, large-conductance channel that is closed under physiological conditions. Opening of the mPTP, leading to abolition of mitochondrial functions, is a major mechanism of myocyte necrosis by ischaemia/reperfusion, and direct inhibition of mPTP opening by use of pharmacological or genetic manipulations limits infarct size in vivo. Multiple pro-survival signal pathways commonly target the mPTP and inhibit its opening. Although the molecular structure of the mPTP has not been established, recent studies have characterized roles of each mPTP subunit and functions of several proteins directly interacting with the mPTP. This article briefly describes the understanding of mPTP regulation and interaction of the mPTP with four proteins (hexokinase II, glycogen synthase kinase-3β, signal transducer and activator of transcription 3, and sirtuin 3) that are downstream of signal pathways relevant to protection from ischaemia/reperfusion injury.
Collapse
Affiliation(s)
- Tetsuji Miura
- Second Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | | |
Collapse
|
37
|
Sanada S, Komuro I, Kitakaze M. Pathophysiology of myocardial reperfusion injury: preconditioning, postconditioning, and translational aspects of protective measures. Am J Physiol Heart Circ Physiol 2011; 301:H1723-41. [PMID: 21856909 DOI: 10.1152/ajpheart.00553.2011] [Citation(s) in RCA: 260] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heart diseases due to myocardial ischemia, such as myocardial infarction or ischemic heart failure, are major causes of death in developed countries, and their number is unfortunately still growing. Preliminary exploration into the pathophysiology of ischemia-reperfusion injury, together with the accumulation of clinical evidence, led to the discovery of ischemic preconditioning, which has been the main hypothesis for over three decades for how ischemia-reperfusion injury can be attenuated. The subcellular pathophysiological mechanism of ischemia-reperfusion injury and preconditioning-induced cardioprotection is not well understood, but extensive research into components, including autacoids, ion channels, receptors, subcellular signaling cascades, and mitochondrial modulators, as well as strategies for modulating these components, has made evolutional progress. Owing to the accumulation of both basic and clinical evidence, the idea of ischemic postconditioning with a cardioprotective potential has been discovered and established, making it possible to apply this knowledge in the clinical setting after ischemia-reperfusion insult. Another a great outcome has been the launch of translational studies that apply basic findings for manipulating ischemia-reperfusion injury into practical clinical treatments against ischemic heart diseases. In this review, we discuss the current findings regarding the fundamental pathophysiological mechanisms of ischemia-reperfusion injury, the associated protective mechanisms of ischemic pre- and postconditioning, and the potential seeds for molecular, pharmacological, or mechanical treatments against ischemia-reperfusion injury, as well as subsequent adverse outcomes by modulation of subcellular signaling mechanisms (especially mitochondrial function). We also review emerging translational clinical trials and the subsistent clinical comorbidities that need to be overcome to make these trials applicable in clinical medicine.
Collapse
Affiliation(s)
- Shoji Sanada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | |
Collapse
|
38
|
Shintani-Ishida K, Yoshida KI. Ischemia induces phospholamban dephosphorylation via activation of calcineurin, PKC-α, and protein phosphatase 1, thereby inducing calcium overload in reperfusion. Biochim Biophys Acta Mol Basis Dis 2011; 1812:743-51. [DOI: 10.1016/j.bbadis.2011.03.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 02/23/2011] [Accepted: 03/21/2011] [Indexed: 11/30/2022]
|
39
|
|
40
|
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
|
41
|
Lovell MJ, Yasin M, Lee KL, Cheung KK, Shintani Y, Collino M, Sivarajah A, Leung KY, Takahashi K, Kapoor A, Yaqoob MM, Suzuki K, Lythgoe MF, Martin J, Munroe PB, Thiemermann C, Mathur A. Bone marrow mononuclear cells reduce myocardial reperfusion injury by activating the PI3K/Akt survival pathway. Atherosclerosis 2010; 213:67-76. [DOI: 10.1016/j.atherosclerosis.2010.07.045] [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] [Received: 01/13/2010] [Revised: 07/22/2010] [Accepted: 07/23/2010] [Indexed: 11/16/2022]
|
42
|
Abstract
Infarct size is determined not only by the severity of ischemia but also by pathological processes initiated at reperfusion. Accumulating experimental evidence indicates that lethal reperfusion injury might account for up to half of the final size of the myocardial infarct. Ischemic postconditioning (brief repeated periods of ischemia-reperfusion applied at the onset of coronary reflow) has been recently described as a powerful cardioprotection mechanism that prevents lethal reperfusion injury. This is the first method proven to reduce the final infarct size by about 50% in several in vivo models and to be confirmed in recent preliminary human studies. The molecular pathways are incompletely mapped but they probably converge to a mitochondrial key target: the mitochondrial permeability transition pore (PTP) which opening during early reperfusion is an event that promotes myocardial cell death. In different animal models and experimental settings, pharmacological PTP inhibition at the onset of reperfusion reproduces all the cardioprotective effects of ischemic postconditioning. In a recent proof-of-concept trial, the administration (just before percutaneous coronary intervention) of cyclosporine A, a potent PTP inhibitor, was associated with smaller infarct size. This review will focus on the physiological preclinical data on both ischemic and pharmacological postconditioning that are relevant to their translation to clinical therapeutics.
Collapse
|
43
|
Brar BK, Helgeland E, Mahata SK, Zhang K, O'Connor DT, Helle KB, Jonassen AK. Human catestatin peptides differentially regulate infarct size in the ischemic-reperfused rat heart. ACTA ACUST UNITED AC 2010; 165:63-70. [PMID: 20655339 DOI: 10.1016/j.regpep.2010.07.153] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 06/29/2010] [Accepted: 07/08/2010] [Indexed: 01/06/2023]
Abstract
In acute myocardial infarction increased plasma levels of chromogranin A are correlated with decreased survival. At the human chromogranin A gene locus there are two naturally occurring amino acid substitution variants within the catestatin region, i.e. Gly³⁶⁴Ser and Pro³⁷⁰Leu, displaying differential potencies towards inhibition of nicotinic cholinergic agonist-evoked catecholamine secretion from sympathochromaffin cells and different degrees of processing from the prohormone. Here, we examine whether two of the variants and the wild type catestatin may affect the development of infarct size during ischemic reperfusion in the Langendorff rat heart model. The hearts were subjected to regional ischemia followed by reperfusion in the presence or absence of synthetic variants of human catestatin. Compared to the Gly³⁶⁴Ser variant both the wild type and Pro³⁷⁰Leu variants increased infarct size while decreasing the cardiac levels of phosphorylated Akt and two of its downstream targets, FoxO1 and BAD. In conclusion, these findings suggest that, in contrast to the Gly³⁶⁴Ser variant, wild type catestatin and the Pro³⁷⁰Leu variant (allele frequency ~0.3%) increased myocardial infarct size via a mechanism involving dephosphorylation of Akt and the two downstream targets during ischemic reperfusion in the isolated rat heart.
Collapse
|
44
|
Bell RM, Yellon DM. There is More to Life than Revascularization: Therapeutic Targeting of Myocardial Ischemia/Reperfusion Injury. Cardiovasc Ther 2010; 29:e67-79. [DOI: 10.1111/j.1755-5922.2010.00190.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
45
|
Zhu J, Rebecchi MJ, Tan M, Glass PSA, Brink PR, Liu L. Age-associated differences in activation of Akt/GSK-3beta signaling pathways and inhibition of mitochondrial permeability transition pore opening in the rat heart. J Gerontol A Biol Sci Med Sci 2010; 65:611-9. [PMID: 20427381 DOI: 10.1093/gerona/glq035] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pretreatment with isoflurane decreased myocardial infarction size in young rats (3-5 months) but not in old rats (20-24 months). To understand the mechanisms underlying the failure to protect the old myocardium, differences in phosphorylation of Akt/GSK-3beta and age-associated differences in mitochondrial permeability transition pore (mPTP) opening in the aging heart in vivo were measured. Isoflurane significantly increased Akt and GSK-3beta phosphorylation in the young groups. In contrast, levels of p-Akt and p-GSK-3beta were highly elevated in the old sham control groups. Isoflurane preconditioning significantly reduced the fall in NAD(+) levels induced by ischemia/reperfusion injury in the young animals, reflecting the inhibition of mPTP opening. In the old animals, however, isoflurane failed to prevent the fall in NAD(+) levels induced by ischemia/reperfusion injury. Lack of isoflurane-induced cardioprotective effects, seen in the old animals, can be explained by age-related differences in Akt/GSK-3beta signaling pathway and the inability to reduce mPTP opening following ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Jiang Zhu
- Department of Anesthesiology, Stony Brook University School of Medicine, HSC L4 060, Stony Brook, NY 11794, USA
| | | | | | | | | | | |
Collapse
|
46
|
Grundmann S, Bode C, Moser M. Mannose-binding lectin: an ancient molecule with new implications in myocardial infarction. Eur Heart J 2010; 31:1163-4. [PMID: 20233793 DOI: 10.1093/eurheartj/ehq070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
47
|
Saeed M, Hetts S, Wilson M. Reperfusion injury components and manifestations determined by cardiovascular MR and MDCT imaging. World J Radiol 2010; 2:1-14. [PMID: 21160735 PMCID: PMC2999314 DOI: 10.4329/wjr.v2.i1.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 01/06/2010] [Accepted: 01/13/2010] [Indexed: 02/06/2023] Open
Abstract
Advances in magnetic resonance (MR) and computed tomography (CT) imaging have improved visualization of acute and scar infarct. Over the past decade, there have been and continues to be many significant technical advancements in cardiac MR and multi-detector computed tomography (MDCT) technologies. The strength of MR imaging relies on a variety of pulse sequences and the ability to noninvasively provide information on myocardial structure, function and perfusion in a single imaging session. The recent technical developments may also allow CT technologies to rise to the forefront for evaluating clinical ischemic heart disease. Components of reperfusion injury including myocardial edema, hemorrhage, calcium deposition and microvascular obstruction (MO) have been demonstrated using MR and CT technologies. MR imaging can be used serially and noninvasively in assessing acute and chronic consequences of reperfusion injury because there is no radiation exposure or administration of radioactive materials. MDCT is better suited for assessing coronary artery stenosis and as an alternative technique for assessing viability in patients where MR imaging is contraindicated. Changes in left ventricular (LV) volumes and function measured on cine MR are directly related to infarct size measured on delayed contrast enhanced images. Recent MR studies found that transmural infarct, MO and peri-infarct zone are excellent predictors of poor post-infarct recovery and mortality. Recent MR studies provided ample evidence that growth factor genes and stem cells delivered locally have beneficial effects on myocardial viability, perfusion and function. The significance of deposited calcium in acute infarct detected on MDCT requires further studies. Cardiac MR and MDCT imaging have the potential for assessing reperfusion injury components and manifestations.
Collapse
|
48
|
Kim JH. Protective Roles of Ginseng Saponin in Cardiac Ischemia and Reperfusion Injury. J Ginseng Res 2009. [DOI: 10.5142/jgr.2009.33.4.283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
Mani SK, Balasubramanian S, Zavadzkas JA, Jeffords LB, Rivers WT, Zile MR, Mukherjee R, Spinale FG, Kuppuswamy D. Calpain inhibition preserves myocardial structure and function following myocardial infarction. Am J Physiol Heart Circ Physiol 2009; 297:H1744-51. [PMID: 19734364 PMCID: PMC2781387 DOI: 10.1152/ajpheart.00338.2009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 09/01/2009] [Indexed: 12/13/2022]
Abstract
Cardiac pathology, such as myocardial infarction (MI), activates intracellular proteases that often trigger programmed cell death and contribute to maladaptive changes in myocardial structure and function. To test whether inhibition of calpain, a Ca(2+)-dependent cysteine protease, would prevent these changes, we used a mouse MI model. Calpeptin, an aldehydic inhibitor of calpain, was intravenously administered at 0.5 mg/kg body wt before MI induction and then at the same dose subcutaneously once per day. Both calpeptin-treated (n = 6) and untreated (n = 6) MI mice were used to study changes in myocardial structure and function after 4 days of MI, where end-diastolic volume (EDV) and left ventricular ejection fraction (EF) were measured by echocardiography. Calpain activation and programmed cell death were measured by immunohistochemistry, Western blotting, and TdT-mediated dUTP nick-end labeling (TUNEL). In MI mice, calpeptin treatment resulted in a significant improvement in EF [EF decreased from 67 + or - 2% pre-MI to 30 + or - 4% with MI only vs. 41 + or - 2% with MI + calpeptin] and attenuated the increase in EDV [EDV increased from 42 + or - 2 microl pre-MI to 73 + or - 4 microl with MI only vs. 55 + or - 4 microl with MI + calpeptin]. Furthermore, calpeptin treatment resulted in marked reduction in calpain- and caspase-3-associated changes and TUNEL staining. These studies indicate that calpain contributes to MI-induced alterations in myocardial structure and function and that it could be a potential therapeutic target in treating MI patients.
Collapse
Affiliation(s)
- Santhosh K Mani
- Division of Cardiology, Department of Medicine, Charleston, South Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pomblum VJ, Korbmacher B, Cleveland S, Sunderdiek U, Klocke RC, Schipke JD. Cardiac stunning in the clinic: the full picture. Interact Cardiovasc Thorac Surg 2009; 10:86-91. [PMID: 19773228 DOI: 10.1510/icvts.2009.205666] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cardiac stunning refers to different dysfunctional levels occurring after an episode of acute ischemia, despite blood flow is near normal or normal. The phenomenon was initially identified in animal models, where it has been very well characterized. After being established in the experimental setting, it remained unclear, whether a similar syndrome occurs in humans. In addition, it remained controversial, whether stunning was of any clinical relevance as it is spontaneously reversible. Hence, many studies continue to focus on the properties and mechanisms of stunning, although therapies seem more relevant for attenuating and treating myocardial ischemia/reperfusion (I/R) injury, i.e. to bridge until recovery. This article reviews the different facets of cardiac stunning, i.e. myocardial, vascular/microvascular/endothelial, metabolic, neural/neuronal, and electrical stunning. This review also displays where these facets exist and which clinical relevance they might have. Particular attention is directed to the different therapeutic interventions that the various facets of this I/R-induced cardiac injury might require. A final outlook considers possible alternatives to further reduce the detrimental consequences of brief episodes of ischemia and reperfusion.
Collapse
Affiliation(s)
- Valdeci J Pomblum
- Department of Internal Medicine, Federal University of Santa Maria, Santa Maria (RS), Brazil
| | | | | | | | | | | |
Collapse
|