1
|
de Paula LJC, Uchida AH, Rezende PC, Soares P, Scudeler TL. Protective or Inhibitory Effect of Pharmacological Therapy on Cardiac Ischemic Preconditioning: A Literature Review. Curr Vasc Pharmacol 2022; 20:409-428. [PMID: 35986546 DOI: 10.2174/1570161120666220819163025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 01/25/2023]
Abstract
Ischemic preconditioning (IP) is an innate phenomenon, triggered by brief, non-lethal cycles of ischemia/reperfusion applied to a tissue or organ that confers tolerance to a subsequent more prolonged ischemic event. Once started, it can reduce the severity of myocardial ischemia associated with some clinical situations, such as percutaneous coronary intervention (PCI) and intermittent aortic clamping during coronary artery bypass graft surgery (CABG). Although the mechanisms underlying IP have not been completely elucidated, several studies have shown that this phenomenon involves the participation of cell triggers, intracellular signaling pathways, and end-effectors. Understanding this mechanism enables the development of preconditioning mimetic agents. It is known that a range of medications that activate the signaling cascades at different cellular levels can interfere with both the stimulation and the blockade of IP. Investigations of signaling pathways underlying ischemic conditioning have identified a number of therapeutic targets for pharmacological manipulation. This review aims to present and discuss the effects of several medications on myocardial IP.
Collapse
Affiliation(s)
| | | | - Paulo Cury Rezende
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Paulo Soares
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thiago Luis Scudeler
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Honokiol Provides Cardioprotection from Myocardial Ischemia/Reperfusion Injury (MI/RI) by Inhibiting Mitochondrial Apoptosis via the PI3K/AKT Signaling Pathway. Cardiovasc Ther 2022; 2022:1001692. [PMID: 35414825 PMCID: PMC8977331 DOI: 10.1155/2022/1001692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background Myocardial injury refers to a major complication that occurs in myocardial ischemia/reperfusion injury (MI/RI). Honokiol is a well-recognized active compound extracted from the traditional Chinese herb known as Magnolia officinalis and is utilized in treating different vascular diseases. This research is aimed at examining whether Honokiol might alleviate myocardial injury in an MI/RI model. Methods Seventy-eight male C57BL/6 mice were categorized randomly into three cohorts including the Sham operation (Sham) cohort, the MI/RI cohort (Con), and the Honokiol cohort (n = 26 for each cohort). The mice in the Honokiol cohort were treated with Honokiol before MI/RI surgery (0.2 mg/kg/day for 14 days, intraperitoneal), while the mice in the Con cohort were given an intraperitoneal injection with an equivalent volume of vehicle (DMSO) daily in 14 days prior to exposure to MI/RI. After the surgery, creatine kinase- (CK-) MB and cardiac troponin T (cTnT) levels, as well as the infarct area, were measured to assess the degree of myocardial damage. Apoptotic levels were detected using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining. Electron microscopy was utilized to identify mitochondrial damage. Lastly, the expression levels of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), cleaved caspase-9, cytochrome C (Cyt-C), B cell lymphoma/leukemia-2 (Bcl-2), B cell lymphoma/leukemia-2 associated X (Bax), AKT, p-AKT, PI3K, and p-PI3K were analyzed utilizing western blotting. Results Honokiol can reduce the MI/RI-induced cTnT and CK-MB levels, apoptosis index, and mitochondrial swelling in cardiomyocytes via activating the PI3K/AKT signaling pathway. Conclusion Honokiol provides cardiac protection from MI/RI by suppressing mitochondrial apoptosis through the PI3K/AKT signaling pathway.
Collapse
|
3
|
Zare gashti R, Mohammadi H. Sodium dithionate (Na2S2O4) induces oxidative damage in mice mitochondria heart tissue. Toxicol Rep 2022; 9:1391-1397. [DOI: 10.1016/j.toxrep.2022.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 10/17/2022] Open
|
4
|
Chen XY, Wang JQ, Cheng SJ, Wang Y, Deng MY, Yu T, Wang HY, Zhou WJ. Diazoxide Post-conditioning Activates the HIF-1/HRE Pathway to Induce Myocardial Protection in Hypoxic/Reoxygenated Cardiomyocytes. Front Cardiovasc Med 2021; 8:711465. [PMID: 34938777 PMCID: PMC8687117 DOI: 10.3389/fcvm.2021.711465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Background: Previous studies have shown that diazoxide can protect against myocardial ischemia-reperfusion injury (MIRI). The intranuclear hypoxia-inducible factor-1 (HIF-1)/hypoxia-response element (HRE) pathway has been shown to withstand cellular damage caused by MIRI. It remains unclear whether diazoxide post-conditioning is correlated with the HIF-1/HRE pathway in protective effect on cardiomyocytes. Methods: An isolated cardiomyocyte model of hypoxia-reoxygenation injury was established. Prior to reoxygenation, cardiomyocytes underwent post-conditioning treatment by diazoxide, and 5-hydroxydecanoate (5-HD), N-(2-mercaptopropionyl)-glycine (MPG), or dimethyloxallyl glycine (DMOG) followed by diazoxide. At the end of reoxygenation, ultrastructural morphology; mitochondrial membrane potential; interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), reactive oxygen species (ROS), and HIF-1α levels; and downstream gene mRNA and protein levels were analyzed to elucidate the protective mechanism of diazoxide post-conditioning. Results: Diazoxide post-conditioning enabled activation of the HIF-1/HRE pathway to induce myocardial protection. When the mitoKATP channel was inhibited and ROS cleared, the diazoxide effect was eliminated. DMOG was able to reverse the effect of ROS absence to restore the diazoxide effect. MitoKATP and ROS in the early reoxygenation phase were key to activation of the HIF-1/HRE pathway. Conclusion: Diazoxide post-conditioning promotes opening of the mitoKATP channel to generate a moderate ROS level that activates the HIF-1/HRE pathway and subsequently induces myocardial protection.
Collapse
Affiliation(s)
- Xi-Yuan Chen
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
- Department of Anesthesiology, The Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Jia-Qi Wang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Si-Jing Cheng
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Yan Wang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Meng-Yuan Deng
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Hai-Ying Wang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Wen-Jing Zhou
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| |
Collapse
|
5
|
Abstract
The ischemia-reperfusion syndrome complicates the course of a number of emergency conditions in various fields of clinical medicine, determines the course, prognosis and outcome of the disease. This review examines various aspects of the etiology, pathogenesis, and clinical manifestations of this syndrome. Particular attention is paid to its prevention and treatment. It is indicated that most of the studies devoted to this problem are of an experimental nature. The use of preparations based on succinic acid in the clinic is seen as the most promising direction in solving this issue.
Collapse
Affiliation(s)
- M I Neimark
- Altai State Medical University, Barnaul, Russia
| |
Collapse
|
6
|
|
7
|
Baig S, Moyle B, Nair KPS, Redgrave J, Majid A, Ali A. Remote ischaemic conditioning for stroke: unanswered questions and future directions. Stroke Vasc Neurol 2021; 6:298-309. [PMID: 33903181 PMCID: PMC8258051 DOI: 10.1136/svn-2020-000722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/13/2021] [Accepted: 01/31/2021] [Indexed: 11/07/2022] Open
Abstract
Remote ischaemic conditioning (RIC) refers to a process whereby periods of intermittent ischaemia, typically via the cyclical application of a blood pressure cuff to a limb at above systolic pressure, confers systemic protection against ischaemia in spatially distinct vascular territories. The mechanisms underlying this have not been characterised fully but have been shown to involve neural, hormonal and systemic inflammatory signalling cascades. Preclinical and early clinical studies have been promising and suggest beneficial effects of RIC in acute ischaemic stroke, symptomatic intracranial stenosis and vascular cognitive impairment. Through systematic searches of several clinical trials databases we identified 48 active clinical trials of RIC in ischaemic stroke, intracerebral haemorrhage and subarachnoid haemorrhage. We summarise the different RIC protocols and outcome measures studied in ongoing clinical trials and highlight which studies are most likely to elucidate the underlying biological mechanisms of RIC and characterise its efficacy in the near future. We discuss the uncertainties of RIC including the optimal frequency and duration of therapy, target patient groups, cost-effectiveness, the confounding impact of medications and the absence of a clinically meaningful biomarker of the conditioning response. With several large clinical trials of RIC expected to report their outcomes within the next 2 years, this review aims to highlight the most important studies and unanswered questions that will need to be addressed before this potentially widely accessible and low-cost intervention can be used in clinical practice.
Collapse
Affiliation(s)
- Sheharyar Baig
- Cerebrovascular Medicine, The University of Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - Bethany Moyle
- Cerebrovascular Medicine, The University of Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | | | - Jessica Redgrave
- Cerebrovascular Medicine, The University of Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - Arshad Majid
- Faculty of Medicine and Dentistry, University of Sheffield, Sheffield, UK
| | - Ali Ali
- Geriatrics and Stroke Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK .,Sheffield NIHR Biomedical Research Centre, The University of Sheffield, Sheffield, UK
| |
Collapse
|
8
|
de Miranda DC, de Oliveira Faria G, Hermidorff MM, Dos Santos Silva FC, de Assis LVM, Isoldi MC. Pre- and Post-Conditioning of the Heart: An Overview of Cardioprotective Signaling Pathways. Curr Vasc Pharmacol 2020; 19:499-524. [PMID: 33222675 DOI: 10.2174/1570161119666201120160619] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
Since the discovery of ischemic pre- and post-conditioning, more than 30 years ago, the knowledge about the mechanisms and signaling pathways involved in these processes has significantly increased. In clinical practice, on the other hand, such advancement has yet to be seen. This article provides an overview of ischemic pre-, post-, remote, and pharmacological conditioning related to the heart. In addition, we reviewed the cardioprotective signaling pathways and therapeutic agents involved in the above-mentioned processes, aiming to provide a comprehensive evaluation of the advancements in the field. The advancements made over the last decades cannot be ignored and with the exponential growth in techniques and applications. The future of pre- and post-conditioning is promising.
Collapse
Affiliation(s)
- Denise Coutinho de Miranda
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Gabriela de Oliveira Faria
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Milla Marques Hermidorff
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Fernanda Cacilda Dos Santos Silva
- Laboratory of Cardiovascular Physiology, Department of Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Leonardo Vinícius Monteiro de Assis
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Mauro César Isoldi
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
9
|
Ginkgetin Alleviates Inflammation, Oxidative Stress, and Apoptosis Induced by Hypoxia/Reoxygenation in H9C2 Cells via Caspase-3 Dependent Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1928410. [PMID: 33204684 PMCID: PMC7661124 DOI: 10.1155/2020/1928410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/04/2020] [Accepted: 08/24/2020] [Indexed: 11/18/2022]
Abstract
Ginkgetin, the extract of Ginkgo biloba leaves, has been reported to exert preventive and therapeutic effects on cardiovascular disease. However, little is known about its role in myocardial ischemia-reperfusion injury (MIRI). The present study aimed to unveil the function of ginkgetin in cardiomyocytes subjected to hypoxia/reoxygenation (H/R) injury. Cell Counting Kit-8 (CCK-8) was employed to evaluate the impact of ginkgetin on cell viability in the absence or presence of H/R. Proinflammatory cytokines and malondialdehyde (MDA), reactive oxygen species (SOD), and lactate dehydrogenase (LDH) were determined via corresponding kits. In addition, flow cytometry was performed to detect apoptotic level. Western blot analysis was utilized to estimate caspase-3 and cytochrome C. Ginkgetin had no significant effect on cell viability; however, it could enhance viability of H9C2 cells exposed to H/R. Inflammation and oxidative stress induced by H/R injury were relieved via pretreatment with ginkgetin. Preconditioning of ginkgetin also decreased apoptotic rate and the protein levels of caspase-3, cytochrome C under H/R condition. Furthermore, 2-HBA, an inducer of caspase-3, was used for the activation of caspase-3 signaling pathway. It was found that induction of caspase-3 eliminated the protective effect of ginkgetin on H9C2 cells exposed to H/R. These results indicated that ginkgetin attenuated inflammation, oxidative stress, and apoptosis. These protective roles of ginkgetin may attribute to caspase-3 dependent pathway.
Collapse
|
10
|
Wang J, Liu J, Xie L, Cai X, Ma X, Gong J. Bisoprolol, a β 1 antagonist, protects myocardial cells from ischemia-reperfusion injury via PI3K/AKT/GSK3β pathway. Fundam Clin Pharmacol 2020; 34:708-720. [PMID: 32363697 DOI: 10.1111/fcp.12562] [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: 03/04/2020] [Revised: 04/23/2020] [Accepted: 04/29/2020] [Indexed: 12/30/2022]
Abstract
The aim of this work was to explore whether bisoprolol plays a protective role in cardiomyocytes against ischemia-reperfusion injury via PI3K/AKT/ GSK3β pathway. We pretreated male Sprague Dawley (SD) rats with bisoprolol by oral administration prior to 0.5 h ischemia/4 h reperfusion. Myocardial infarct size and serum levels of cTnI and CK-MB were measured. In vitro, H9c2 cells were treated with hypoxia and reoxygenation, followed by measurement of cell viability, apoptosis, ROS production, cytometry, activities of AKT, GSK3β, and p-38 in the presence and absence of GSK3β siRNA. We found that bisoprolol reduced infarct size from 44% in I/R group to 31% in treated group (P < 0.05). The levels of cTnI and CK-MB were decreased from 286 ± 7 pg/mL and 32.2 ± 2 ng/mL in I/R group to 196 ± 2 pg/mL and 19.6 ± 0.9 ng/mL in the treated group, respectively (P < 0.05). Bisoprolol also increased cell viability while decreased apoptosis and ROS production in the treatment of hypoxia/ reoxygenation. Furthermore, bisoprolol increased AKT and GSK3β phosphorylation, an effect that was immediately eliminated by LY294002. GSK3β-specific siRNA experiment further confirmed that bisoprolol protected the myocardium against hypoxia/reoxygenation-induced injury via suppressing GSK3β activity. In conclusion, bisoprolol protected myocardium against ischemia-reperfusion injury via the PI3K/AKT/ GSK3β pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Jing Liu
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Liang Xie
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Xiaomin Cai
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Xiaohua Ma
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Jianbin Gong
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| |
Collapse
|
11
|
Apigenin protects against ischemia-/hypoxia-induced myocardial injury by mediating pyroptosis and apoptosis. In Vitro Cell Dev Biol Anim 2020; 56:307-312. [PMID: 32406012 DOI: 10.1007/s11626-020-00434-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/06/2020] [Indexed: 10/24/2022]
Abstract
Apigenin is a traditional Chinese medicine found in many plants that plays critical roles in several diseases, including cardiovascular diseases. We investigated the protective effect of apigenin against ischemia/hypoxia (I/H)-induced myocardium injury in vitro and explored the potential molecular mechanisms. Together with the flow cytometry results, our results indicated that apigenin attenuated the pyroptosis and apoptosis of myoblastic H9c2 cells that were induced by I/H injury. Furthermore, the pro-inflammatory cytokines (interleukin 18, IL-18, and IL-1β) were investigated for the roles of apigenin in I/H-induced myocardium injury. It was observed that increases in IL-1β and IL-18 levels were significantly reduced by apigenin treatment of I/H-induced H9C2 cells. The results demonstrated that apigenin protected H9c2 cells against I/H-induced myocardium injury. The protective effects were most likely related to the reduction of pyroptosis, apoptosis, and pro-inflammatory cytokines.
Collapse
|
12
|
Oleaga C, Jalilvand G, Legters G, Martin C, Ekman G, McAleer CW, Long CJ, Hickman JJ. A human in vitro platform for the evaluation of pharmacology strategies in cardiac ischemia. APL Bioeng 2019; 3:036103. [PMID: 31431939 PMCID: PMC6692160 DOI: 10.1063/1.5089237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/22/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac ischemic events increase the risk for arrhythmia, heart attack, heart failure, and death and are the leading mortality condition globally. Reperfusion therapy is the first line of treatment for this condition, and although it significantly reduces mortality, cardiac ischemia remains a significant threat. New therapeutic strategies are under investigation to improve the ischemia survival rate; however, the current preclinical models to validate these fail to predict the human outcome. We report the development of a functional human cardiac in vitro system for the study of conduction velocity under ischemic conditions. The system is a bioMEMs platform formed by human iPSC derived cardiomyocytes patterned on microelectrode arrays and maintained in serum-free conditions. Electrical activity changes of conduction velocity, beat frequency, and QT interval (the QT-interval measures the period from onset of depolarization to the completion of repolarization) or action potential length can be evaluated over time and under the stress of ischemia. The optimized protocol induces >80% reduction in conduction velocity, after a 4 h depletion period, and a partial recovery after 72 h of oxygen and nutrient reintroduction. The sensitivity of the platform for pharmacological interventions was challenged with a gap junction modulator (ZP1609), known to prevent or delay the depression of conduction velocity induced by ischemic metabolic stress. ZP1609 significantly improved the drastic drop in conduction velocity and enabled a greater recovery. This model represents a new preclinical platform for studying cardiac ischemia with human cells, which does not rely on biomarker analysis and has the potential for screening novel cardioprotective drugs with readouts that are closer to the measured clinical parameters.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Golareh Jalilvand
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Gregg Legters
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Gail Ekman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | | | | | - James J. Hickman
- Author to whom correspondence should be addressed:. Tel.: +1 407-823-1925
| |
Collapse
|
13
|
Liu DW, Zhang YN, Hu HJ, Zhang PQ, Cui W. Downregulation of microRNA‑199a‑5p attenuates hypoxia/reoxygenation‑induced cytotoxicity in cardiomyocytes by targeting the HIF‑1α‑GSK3β‑mPTP axis. Mol Med Rep 2019; 19:5335-5344. [PMID: 31059047 PMCID: PMC6522876 DOI: 10.3892/mmr.2019.10197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRs) have been identified as critical regulatory molecules in myocardial ischemia/reperfusion injury; however, the exact expression profile of miR-199a-5p in reperfusion injury and the underlying pathogenic mechanisms remain unclear. In the present study, it was revealed that miR-199a-5p expression was significantly increased in the plasma of patients with acute myocardial infarction and in a H9c2 cell model of oxygen-glucose deprivation and reperfusion (OGD/R) via reverse transcription-quantitative PCR. H9c2 cells were transfected with miR-199a-5p mimic or inhibitor, or short interfering RNA (siRNA) specific to hypoxia-inducible factor-1α (HIF-1α). MTS, lactate dehydrogenase (LDH), TUNEL staining and flow cytometry assays were performed to determine the proliferation, LDH activity, apoptosis and mitochondrial membrane potential (ΔΨm) of H9c2 cells, respectively. The overexpression of miR-199a-5p in the OGD/R cell model significantly decreased the viability and increased the lactate dehydrogenase leakage of cells; whereas knockdown of miR-199-5p induced the opposing effects. Additionally, inhibition of miR-199-5p significantly attenuated OGD/R-induced alterations to the mitochondrial transmembrane potential (ΔΨm) and increases in the apoptosis of cells. Furthermore, the overexpression or knockdown of miR-199a-5p decreased or increased the expression of HIF-1α and phosphorylation of glycogen synthase kinase 3β (GSK3β) in OGD/R-treated H9c2 cells. Additionally, siRNA-mediated downregulation of HIF-1α decreased phosphorylated (p)-GSK3β (Ser9) levels and reversed the protective effects of miR-199a-5p inhibition on OGD/R-injured H9c2 cells. Similarly, treatment with LiCl (a specific inhibitor of p-GSK3β) also attenuated the protective effects of miR-199a-5p knockdown on OGD/R-injured H9c2 cells. Mechanistic studies revealed that HIF-1α was a target of miR-199a-5p, and that HIF-1α downregulation suppressed the expression of p-GSK3β in OGD/R-injured H9c2 cells. Furthermore, an miR-199a-5p inhibitor increased the interaction between p-GSK3β and adenine nucleotide transferase (ANT), which was decreased by OGD/R. Additionally, miR-199a-5p inhibitor reduced the OGD/R-induced interaction between ANT and cyclophilin D (Cyp-D), potentially leading to the increased mitochondrial membrane potential in inhibitor-transfected OGD/R-injured H9c2 cells. Collectively, the present study identified a novel regulatory pathway in which the upregulation of miR-199a-5p reduced the expression of HIF-1α and p-GSK3β, and potentially suppresses the interaction between p-GSK3β and ANT, thus promoting the interaction between ANT and Cyp-D and potentially inducing cytotoxicity in OGD/R-injured H9c2 cells.
Collapse
Affiliation(s)
- Da-Wei Liu
- Department of Cardiology, Second Hospital of Hebei Medical University and Hebei Institute of Cardiovascular Research, Shijiazhuang, Hebei 050011, P.R. China
| | - Ya-Nan Zhang
- Department of Cardiology, Second Hospital of Hebei Medical University and Hebei Institute of Cardiovascular Research, Shijiazhuang, Hebei 050011, P.R. China
| | - Hai-Juan Hu
- Department of Cardiology, Second Hospital of Hebei Medical University and Hebei Institute of Cardiovascular Research, Shijiazhuang, Hebei 050011, P.R. China
| | - Pu-Qiang Zhang
- Department of Cardiology, Second Hospital of Hebei Medical University and Hebei Institute of Cardiovascular Research, Shijiazhuang, Hebei 050011, P.R. China
| | - Wei Cui
- Department of Cardiology, Second Hospital of Hebei Medical University and Hebei Institute of Cardiovascular Research, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
14
|
Wang J, Wu D, Wang H. Hydrogen sulfide plays an important protective role by influencing autophagy in diseases. Physiol Res 2019; 68:335-345. [PMID: 30904008 DOI: 10.33549/physiolres.933996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Autophagy can regulate cell growth, proliferation, and stability of cell environment. Its dysfunction can be involved in a variety of diseases. Hydrogen sulfide (H(2)S) is an important signaling molecule that regulates many physiological and pathological processes. Recent studies indicate that H(2)S plays an important protective role in many diseases through influencing autophagy, but its mechanism is not fully understood. This article reviewed the progress about the effect of H(2)S on autophagy in diseases in recent years in order to provide theoretical basis for the further research on the interaction of H(2)S and autophagy and the mechanisms involved.
Collapse
Affiliation(s)
- J Wang
- School of Basic Medical Science, Henan University, Kaifeng, Henan, China.
| | | | | |
Collapse
|
15
|
Liu J, Li Y, Tang Y, Cheng J, Wang J, Li J, Ma X, Zhuang W, Gong J, Liu Z. Rhein protects the myocardiac cells against hypoxia/reoxygention-induced injury by suppressing GSK3β activity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:1-6. [PMID: 30466606 DOI: 10.1016/j.phymed.2018.06.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 05/22/2018] [Accepted: 06/19/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Rhein, an anthraquinone compound isolated from rhubarb, has been shown to protect the pancreatic β cells from hyperglycemia induced apoptosis in our previous studies. PURPOSE In the present study, we examined whether rhein can protect myocardial cells against ischemia reperfusion (I/R)-induced apoptosis and investigated the underlying mechanism. METHODS We used an in vitro model of myocardial hypoxia/reoxygenation (H/R) injury. H9c2 cells were incubated with rhein for 1 h and then subjected to hypoxia for 6 h, followed by reoxygenation for 2 h. Cells viability, apoptosis and ROS were assayed for the treated cells. AKT, p-AKT, GSK3β, p- GSK3β, P38 and p-P38 proteins were analyzed using Western blotting. PI3K/AKT inhibitor, LY294002, and GSK3β siRNA were also used to determine the signaling pathways involved in the protection by rhein. RESULTS Rhein increased viability, decreased apoptosis and ROS production, of the cells that were exposed to H/R. Rhein also increased the phosphorylation of AKT and GSK3β, an effect that was eliminated by LY294002. GSK3β silencing by siRNA showed similar effect as LY294002. The p-P38 level was upregulated by H/R and downregulated in the presence of rhein; however, the p-P38 downregulation was completely abolished by GSK3β silencing. CONCLUSION Rhein protects myocardial H9c2 cells against hypoxia/reoxygenation induced injury via AKT/ GSK3β/p38 pathway.
Collapse
Affiliation(s)
- Jing Liu
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Yanming Li
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Yi Tang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jinghua Cheng
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jing Wang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jianhua Li
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Xiaohua Ma
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Wei Zhuang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jianbin Gong
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China.
| | - Zhihong Liu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
16
|
Besch G, Perrotti A, Salomon du Mont L, Puyraveau M, Ben-Said X, Baltres M, Barrucand B, Flicoteaux G, Vettoretti L, Samain E, Chocron S, Pili-Floury S. Impact of intravenous exenatide infusion for perioperative blood glucose control on myocardial ischemia-reperfusion injuries after coronary artery bypass graft surgery: sub study of the phase II/III ExSTRESS randomized trial. Cardiovasc Diabetol 2018; 17:140. [PMID: 30384842 PMCID: PMC6211400 DOI: 10.1186/s12933-018-0784-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The aim of the study was to investigate whether intravenous (iv) infusion of exenatide, a synthetic GLP-1 receptor agonist, could provide a protective effect against myocardial ischemia-reperfusion injury after coronary artery bypass graft (CABG) surgery. METHODS A sub study analysis of patients > 18 years admitted for elective CABG and included in the ExSTRESS trial was conducted. Patients were randomized to receive either iv exenatide (1-h bolus of 0.05 µg min-1 followed by a constant infusion of 0.025 µg min-1) (exenatide group) or iv insulin therapy (control group) for blood glucose control (target range 100-139 mg dl-1) during the first 48 h after surgical incision. All serum levels of troponin I measured during routine care in the Cardiac Surgery ICU were recorded. The primary outcome was the highest value of plasma concentration of troponin I measured between 12 and 24 h after ICU admission. The proportion of patients presenting an echocardiographic left ventricular ejection fraction (LVEF) > 50% at the follow-up consultation was compared between the two groups. RESULTS Finally, 43 and 49 patients were analyzed in the control and exenatide groups, respectively {age: 69 [61-76] versus 71 [63-75] years; baseline LVEF < 50%: 6 (14%) versus 16 (32%) patients; on-pump surgery: 29 (67%) versus 33 (67%) patients}. The primary outcome did not significantly differ between the two groups (3.34 [1.06-6.19] µg l-1 versus 2.64 [1.29-3.85] µg l-1 in the control and exenatide groups, respectively; mean difference (MD) [95% confidence interval (95% CI)] 0.16 [- 0.25; 0.57], p = 0.54). The highest troponin value measured during the first 72 h in the ICU was 6.34 [1.36-10.90] versus 5.04 [2.39-7.18] µg l-1, in the control and exenatide groups respectively (MD [95% CI] 0.20 [- 0.22; 0.61], p = 0.39). At the follow-up consultation, 5 (12%) versus 8 (16%) patients presented a LVEF < 50% in the control and in the exenatide groups respectively (relative risk [95% CI] 0.68 [0.16; 2.59], p = 0.56). CONCLUSIONS Postoperative iv exenatide did not provide any additional cardioprotective effect compared to iv insulin in low-risk patients undergoing scheduled CABG surgery. Trial registration ClinicalTrials.gov Identifier NCT01969149, date of registration: January 7th, 2015; EudraCT No. 2009-009254-25 A, date of registration: January 6th, 2009.
Collapse
Affiliation(s)
- Guillaume Besch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France.
| | - Andrea Perrotti
- Department of Cardiothoracic Surgery, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Lucie Salomon du Mont
- Department of Vascular Surgery, University Hospital of Besancon, and, EA3920, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Marc Puyraveau
- Clinical Methodology Center, University Hospital of Besancon, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Xavier Ben-Said
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Maude Baltres
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Benoit Barrucand
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Guillaume Flicoteaux
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Lucie Vettoretti
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Emmanuel Samain
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Sidney Chocron
- Department of Cardiothoracic Surgery, University Hospital of Besancon, and, EA3920, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| | - Sebastien Pili-Floury
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Besancon, and, EA3920 and SFR-FED 4234 INSERM, University of Franche-Comte, 3 bvd Alexander Fleming, 25000, Besançon, France
| |
Collapse
|
17
|
Cardioprotective Activity of 2,6-Diisobornyl-4-Methylphenol in Acute Myocardial Ischemia/Reperfusion in Rats. Bull Exp Biol Med 2018; 165:657-659. [PMID: 30225716 DOI: 10.1007/s10517-018-4235-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Indexed: 12/24/2022]
Abstract
We studied the cardioprotective effect of 2,6-diisobornyl-4-methylphenol under conditions of myocardial ischemia/reperfusion in rats. Daily administration of 2,6-diisobornyl-4-methylphenol (100 mg/kg intragastrically) over 3 days before and 5 days after modeling of myocardial ischemia/reperfusion prevented the increase in the infarction area by almost 2 times in comparison with the control by day 5 after recirculation. The type and severity of pathological changes in ECG parameters reflecting necrotic changes in the myocardium under the action of the compound significantly decreased by day 35 of the experiment. Animal survival rate during the first 24 h after ischemia/reperfusion modeling in the experimental group was by 29% higher than in the control group.
Collapse
|
18
|
Abstract
ST-segment elevation myocardial infarction is a major cause of morbidity and mortality worldwide. Reperfusion injury (RI) following the opening of an occluded coronary artery mitigates the effect of reperfusion by further accentuating ischemic damage and increasing infarct size. Experimental studies have shown that nearly 50% of final infarct size is attributable to RI, an elusive phenomenon that remains resistant to treatment. This review proposes a hypothesis to explain the failure of strategies that have been used in an attempt to prevent RI. This hypothesis suggests that, after a certain duration of myocardial ischemia in the affected myocardium, three phases of myocardial damage occur: reversible ischemia, irreversible ischemia, and necrosis. In the reversible ischemia phase, cellular adaptive responses remain functional, and cellular repair and thus recovery of cellular functions is possible, whereas in the irreversible ischemia phase protective maneuvers fail to confer cytoprotection. Preventive therapies for RI fail because they cannot prevent cell death once cells have entered the irreversible ischemia phase, although they may succeed in postponing cell death. Failure to salvage myocardium with irreversible ischemia in addition to postponement and change in the mode of cell death (mainly from necrosis to apoptosis) by various RI preventive strategies may be the key to understanding the failure of these strategies in the clinical setting, despite their success in the reduction of infarct size in the experimental setting. Early reperfusion before large amounts of myocardium at risk reach the stage of irreversible ischemia is the best strategy for reduction of RI-related myocardial damage.
Collapse
|
19
|
Effect of Allicin against Ischemia/Hypoxia-Induced H9c2 Myoblast Apoptosis via eNOS/NO Pathway-Mediated Antioxidant Activity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3207973. [PMID: 29849702 PMCID: PMC5926492 DOI: 10.1155/2018/3207973] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/28/2018] [Accepted: 03/11/2018] [Indexed: 12/15/2022]
Abstract
Allicin (2-propene-1-sulfinothioic acid S-2-propenyl ester, diallyl thiosulfinate) is the main biologically active ingredient in garlic. The present study investigated the protective effect of allicin against cardiomyocyte apoptosis that was induced by ischemia in vitro and the potential molecular mechanisms that were involved in this antiapoptotic effect. The results indicated that allicin increased H9c2 cell activity and attenuated the rate of apoptosis that was induced by ischemia/hypoxia. Intracellular calcium concentrations significantly decreased in the allicin-treated groups. Bax expression significantly decreased, and Bcl-2 expression increased in allicin-treated rats. Nitric oxide blockade significantly inhibited these effects. Allicin also increased the activity of SOD and NO release and decreased MDA levels. Allicin significantly increased the expression of eNOS, Nrf2, and HO-1 proteins. Collectively, these findings demonstrate that allicin protects H9c2 cells against apoptosis, and this protective effect appears to occur via eNOS/NO pathway-mediated antioxidant activity.
Collapse
|
20
|
Shexiang Tongxin Dropping Pill () Protects against Na 2S 2O 4-Induced Hypoxia-Reoxygenation Injury in H9c2 Cells. Chin J Integr Med 2018; 25:439-445. [PMID: 29619747 DOI: 10.1007/s11655-018-2976-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Indexed: 10/17/2022]
Abstract
OBJECTIVES To investigate the protective effects of Shexiang Tongxin Dropping Pill (, STP) on Na2S2O4-induced hypoxia-reoxygenation injury in cardiomyoblast H9c2 cells. METHODS The cell viability and levels of mRNA and protein expression in H9c2 cells were determined following Na2S2O4-induced hypoxia using Hoechst staining, annexin V/propidium iodide (PI) flow cytometry, real-time polymerase chain reaction and Western blot analysis. RESULTS STP pretreatment significantly increased the viability and inhibited aberrant morphological changes in H9c2 cardiomyoblast cells induced by Na2S2O4 treatment (P<0.05). In addition, STP pretreatment attenuated Na2S2O4-induced hypoxic damage, down-regulated the expression of pro-apoptotic Bax, and up-regulated the expression of anti-apoptotic Bcl-2 in H9c2 cells (P<0.05). CONCLUSIONS STP was strongly cardioprotective in hypoxia-reoxygenation injury by preventing hypoxic damage and inhibiting cellular apoptosis. These results further support the use of STP as an effective drug for the treatment of ischemic heart disease.
Collapse
|
21
|
Abstract
Glycogen synthase kinase-3 beta (GSK3β) is principally is a glycogen synthase phosphorylating enzyme that is well known for its role in muscle metabolism. GSK3β is a serine/threonine protein Kinase, which is responsible for several essential roles in mammalian cells. This enzyme is implicated in the pathophysiology of many conditions involved in homeostasis and cellular immigration. GSK3β is involved in several pathways leading to neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Increasing evidence has shown the potential importance of GSK3β in ischemic heart disease and ischemia-reperfusion pathologies. Reperfusion injury may occur in tissues after prolonged ischemia following reperfusion. Reperfusion injury can be life threatening. Reperfusion injury occurs due to a change in ionic homeostasis, excess free radical production, mitochondrial damage and cell death. There are however clear, cardiac-protective signals; although the molecular pathophysiology is not clearly understood. In normal physiology, GSK3β has a critical role in the cytoprotective pathway. However, it`s controversial role in ischemia and ischemia-reperfusion is a topic of current interest. In this review, we have opted to focus on GSK3β interactions with mitochondria in ischemic heart disease and expand on the therapeutic interventions.
Collapse
|
22
|
Eid RA, Alkhateeb MA, Eleawa S, Al-Hashem FH, Al-Shraim M, El-Kott AF, Zaki MSA, Dallak MA, Aldera H. Cardioprotective effect of ghrelin against myocardial infarction-induced left ventricular injury via inhibition of SOCS3 and activation of JAK2/STAT3 signaling. Basic Res Cardiol 2018; 113:13. [PMID: 29392420 DOI: 10.1007/s00395-018-0671-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/22/2018] [Indexed: 11/27/2022]
Abstract
The molecular mechanisms through which ghrelin exerts its cardioprotective effects during cardiac remodeling post-myocardial infarction (MI) are poorly understood. The aim of this study was to investigate whether the cardioprotection mechanisms are mediated by modulation of JAK/STAT signaling and what triggers this modulation. Rats were divided into six groups (n = 12/group): control, sham, sham + ghrelin (100 µg/kg, s.c., daily, starting 1 day post-MI), MI, MI+ ghrelin, and MI+ ghrelin+ AG490, a potent JAK2 inhibitor (5 mg/kg, i.p., daily). All treatments were administered for 3 weeks. Administration of ghrelin to MI rats improved left ventricle (LV) architecture and restored cardiac contraction. In remote non-infarcted areas of MI rats, ghrelin reduced cardiac inflammation and lipid peroxidation and enhanced antioxidant enzymatic activity. In addition, independent of the growth factor/insulin growth factor-1 (GF/IGF-1) axis, ghrelin significantly increased the phosphorylation of JAK2 and Tyr702 and Ser727 residues of STAT3 and inhibited the phosphorylation of JAK1 and Tyr701 and Ser727 residues of STAT1, simultaneously increasing the expression of BCL-2 and decreasing in the expression of BAX, cleaved CASP3, and FAS. This effect coincided with decreased expression of SOCS3. All these beneficial effects of ghrelin, except its inhibitory action on IL-6 expression, were partially and significantly abolished by the co-administration of AG490. In conclusion, the cardioprotective effect of ghrelin against MI-induced LV injury is exerted via activation of JAK2/STAT3 signaling and inhibition of STAT1 signaling. These effects were independent of the GF/IGF-1 axis and could be partially mediated via inhibition of cardiac IL-6.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cardiovascular Agents/administration & dosage
- Disease Models, Animal
- Ghrelin/administration & dosage
- Heart Ventricles/drug effects
- Heart Ventricles/enzymology
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Interleukin-6/metabolism
- Janus Kinase 2/metabolism
- Male
- Myocardial Infarction/drug therapy
- Myocardial Infarction/enzymology
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Rats, Sprague-Dawley
- STAT1 Transcription Factor/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Suppressor of Cytokine Signaling 3 Protein/metabolism
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Refaat A Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia.
| | - Mahmoud A Alkhateeb
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
| | - Samy Eleawa
- College of Health Sciences, Applied Medical Sciences Department, PAAET, Shuwaikh, Kuwait
| | - Fahaid H Al-Hashem
- Department of Physiology, College of Medicine, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Mubarak Al-Shraim
- Department of Pathology, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
| | - Attalla Farag El-Kott
- Department of Biology, College of Science, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Mohammad A Dallak
- Department of Physiology, College of Medicine, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Hussain Aldera
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
| |
Collapse
|
23
|
Zhou T, Prather ER, Garrison DE, Zuo L. Interplay between ROS and Antioxidants during Ischemia-Reperfusion Injuries in Cardiac and Skeletal Muscle. Int J Mol Sci 2018; 19:ijms19020417. [PMID: 29385043 PMCID: PMC5855639 DOI: 10.3390/ijms19020417] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/14/2018] [Accepted: 01/21/2018] [Indexed: 12/16/2022] Open
Abstract
Ischemia reperfusion (IR), present in myocardial infarction or extremity injuries, is a major clinical issue and leads to substantial tissue damage. Molecular mechanisms underlying IR injury in striated muscles involve the production of reactive oxygen species (ROS). Excessive ROS accumulation results in cellular oxidative stress, mitochondrial dysfunction, and initiation of cell death by activation of the mitochondrial permeability transition pore. Elevated ROS levels can also decrease myofibrillar Ca2+ sensitivity, thereby compromising muscle contractile function. Low levels of ROS can act as signaling molecules involved in the protective pathways of ischemic preconditioning (IPC). By scavenging ROS, antioxidant therapies aim to prevent IR injuries with positive treatment outcomes. Novel therapies such as postconditioning and pharmacological interventions that target IPC pathways hold great potential in attenuating IR injuries. Factors such as aging and diabetes could have a significant impact on the severity of IR injuries. The current paper aims to provide a comprehensive review on the multifaceted roles of ROS in IR injuries, with a focus on cardiac and skeletal muscle, as well as recent advancement in ROS-related therapies.
Collapse
Affiliation(s)
- Tingyang Zhou
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Evan R Prather
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Davis E Garrison
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
24
|
Lin S, Chu J, Zhang L, Chen D, Xiao F, Chen H, Lin J, Chen Y, Zhu Y, Peng J. Protective effects of Shexiang Tongxin Dropping Pill on pituitrin‑induced acute myocardial ischemia in rats. Mol Med Rep 2017; 16:3125-3132. [PMID: 28714023 PMCID: PMC5547953 DOI: 10.3892/mmr.2017.6963] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 05/16/2017] [Indexed: 12/12/2022] Open
Abstract
Shexiang Tongxin Dropping Pill (STP) is an established traditional Chinese medicine that is widely used for the treatment of ischemic heart disease (IHD), although its mechanisms remain unclear. The present study investigated the protective effects of STP following pituitrin (PTT)‑induced myocardial ischemia in rats. ST‑segment elevation, blood rheology, and the serum levels of creatine kinase‑MB (CK‑MB) and lactate dehydrogenase (LDH) were measured. Following heart excision, histological analysis using hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick end labeling were performed. The mRNA expression levels of B‑cell lymphoma 2 (Bcl‑2) and Bcl‑2‑associated X protein (Bax) were determined using reverse transcription‑quantitative polymerase chain reaction, and their protein expression was detected using immunohistochemistry. The results demonstrated that STP treatment protected against ST elevation, lowered whole blood viscosity, and reduced the serum levels of CK‑MB and LDH following acute myocardial ischemia. In addition, STP treatment restored the histopathological change following PTT‑induced myocardial ischemia, and resulted in downregulated expression of Bax and upregulated expression of Bcl‑2 in myocardial tissue. The present study demonstrates the cardioprotective ability of STP in a rat model of myocardial ischemic injury, which may be attributed to its anti‑apoptotic properties. The cardioprotective properties of STP require further investigation to determine whether it may be used for the clinical treatment of IHDs.
Collapse
Affiliation(s)
- Shan Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jianfeng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Ling Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Daxin Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Fei Xiao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Hongwei Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Youqin Chen
- Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Yuling Zhu
- Inner Mongolia Conba Pharmaceutical Co., Ltd., Shanghai 201318, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
25
|
Lee SM, Hutchinson M, Saint DA. The role of Toll-like receptor 4 (TLR4) in cardiac ischaemic-reperfusion injury, cardioprotection and preconditioning. Clin Exp Pharmacol Physiol 2017; 43:864-71. [PMID: 27249055 DOI: 10.1111/1440-1681.12602] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/26/2016] [Accepted: 05/30/2016] [Indexed: 01/04/2023]
Abstract
Cardiac ischaemic-reperfusion injury (IRI) remains the primary cause of mortality throughout the developed world. Molecular mechanisms underlying IRI are complex and are often interlinked with each other driving a synergistic response. Toll-like receptor 4 (TLR4), an immunosurveillance receptor, is known to enhance tissue injury during IRI by enhancing the inflammatory response. The release of endogenous components during IRI bind onto TLR4 leading to the activation of multiple signalling kinases. Once this event occurs these proteins are defined as danger associated molecular patterns molecules (DAMPs) or alarmins. Examples include heat shock proteins, high mobility group box one (HMGB1) and extracellular matrix proteins, all of which are involved in IRI. However, literature in the last two decades suggests that transient stimulation of TLR4 may suppress IRI and thus improve cardiac recovery. Furthermore, it remains to be seen what role TLR4 plays during ischaemic-preconditioning where acute bouts of ischaemia, preceding a harmful bout of ischaemic-reperfusion, is cardioprotective. The other question which also needs to be considered is that if transient TLR4 signalling drives a preconditioning response then what are the ligands which drive this? Hence the second part of this review explores the possible TLR4 ligands which may promote cardioprotection against IRI.
Collapse
Affiliation(s)
- Sam Man Lee
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Mark Hutchinson
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Centre for Nanoscale Biophotonics, University of Adelaide, Adelaide, SA, Australia
| | - David A Saint
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
26
|
Hu J, Zhang YX, Wang L, Ding L, Huang GY, Cai GW, Gao S. Protective effects of Xinji'erkang on myocardial infarction induced cardiac injury in mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:338. [PMID: 28651598 PMCID: PMC5485507 DOI: 10.1186/s12906-017-1846-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/20/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Myocardial infarction (MI) is a major risk factor responsible for morbidity and mortality. Xinji'erkang (XJEK) has been clinically used as an effective medication in the treatment of coronary heart disease and myocarditis. The purpose of this study was to investigate the cardioprotective effect of Xinji'erkang on MI mice. METHODS Forty male mice were randomly assigned into four groups as follows (n = 10): sham, model, MI with administration of XJEK and fosinopril for four weeks. At the end of studies, hemodynamic parameters and electrocardiography (ECG) were recorded. Heart and body mass were measured and heart weight/body weight (HW/BW) ratio was calculated as index of hypertrophy. The hypertrophy of heart and aorta was examined using the hematoxylin and eosin (HE) staining, and the collagen deposition was evaluated using Van Gieson (VG) staining. Serum nitric oxide level (NO), superoxide dismutase (SOD) activity and malondialdehyde (MDA) concentration were assayed by colorimetric analysis. The expressions of endothelial NO synthetase (eNOS) expression in serum and cardiac tissues were determined using ELISA assay and immunohistochemistry. Angiotensin II (Ang II) in serum and cardiac tissues was measured using ELISA assay. Besides, tumor necrosis factor-α (TNF-α), interleukin1β (IL-1β) and interleukin10 (IL-10) were observed in cardiac tissues with ELISA assay as well. RESULTS The administration of XJEK significantly improved cardiac dysfunction and abnormal ECG with reduced HW/BW ratio and ameliorated cardiomyocyte hypertrophy and collagen deposition compared to MI, which was partly due to the decreased SOD and increased MDA in serum. Moreover, XJEK treatment also improved endothelial dysfunction (ED) with not only enhanced eNOS activities in serum and cardiac tissues and elevated NO levels in serum, but also decreased Ang II content in serum and cardiac tissues. Finally, protein expressions of pro-inflammation cytokines, TNF-α and IL-1β in the cardiac tissues with XJEK treatment were significantly decreased compared to model. On the contrary, IL-10, an anti-inflammatory cytokine concentrated in cardiac tissues was significantly enhanced compared to model. CONCLUSION Xinji'erkang exerts cardioprotective effect on myocardial infarction in mice, which may be due to the improvement of endothelial dysfunction and the reduction of oxidative stress and inflammation response.
Collapse
Affiliation(s)
- Juan Hu
- 0000 0000 9490 772Xgrid.186775.aDepartment of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032 China
| | - Yong-xue Zhang
- 0000 0000 9490 772Xgrid.186775.aDepartment of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032 China
| | - Li Wang
- 0000 0000 9490 772Xgrid.186775.aDepartment of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032 China
| | - Ling Ding
- 0000 0000 9490 772Xgrid.186775.aDepartment of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032 China
| | - Guang-yao Huang
- 0000 0000 9490 772Xgrid.186775.aDepartment of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032 China
| | - Guo-wei Cai
- 0000 0000 9490 772Xgrid.186775.aDepartment of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032 China
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
27
|
Mu F, Duan J, Bian H, Yin Y, Zhu Y, Wei G, Guan Y, Wang Y, Guo C, Wen A, Yang Y, Xi M. Cardioprotective effects and mechanism of Radix Salviae miltiorrhizae and Lignum Dalbergiae odoriferae on rat myocardial ischemia/reperfusion injury. Mol Med Rep 2017; 16:1759-1770. [PMID: 28656200 PMCID: PMC5562082 DOI: 10.3892/mmr.2017.6821] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 04/25/2017] [Indexed: 01/03/2023] Open
Abstract
Radix Salviae miltiorrhizae (SM) and Lignum Dalbergiae odoriferae (DO) are traditional Chinese medicinal herbs used to treat ischemic heart disease and other cardiovascular diseases; however, to the best of our knowledge, there are currently few studies regarding their effects. The present study aimed to investigate the cardioprotective effects of SM and DO during myocardial ischemia/reperfusion (MI/R) injury in rats, and explore the molecular mechanisms that underlie their actions. In the present study, Sprague-Dawley rats were pretreated with SM, the aqueous extract of DO (DOA) and the volatile oil of DO (DOO), either as a monotherapy or in combination for 7 days. Subsequently, the rats were subjected to 30 min of ischemia followed by 180 min of reperfusion. Traditional pharmacodynamic evaluation and metabonomics based on gas chromatography/time-of-flight mass spectrometry were used to identify the therapeutic effects of these traditional Chinese medicines. The results revealed that SM, DOA and DOO monotherapies ameliorated cardiac function, and this effect was strengthened further when used in combined therapies. Among the combined treatments, SM + DOO exhibited the greatest potential (P<0.05) to improve electrocardiogram results and heart rate, reduce the heart weight index and myocardial infarct size, and decrease the levels of creatine kinase-MB and lactate dehydrogenase. In addition, metabonomics-based findings, including the principal component analysis and partial least squares discriminant analysis score plot of the metabolic state in rat serum, provided confirmation for the aforementioned results, verifying that SM + DOO exerted synergistic therapeutic efficacies to exhibit a greater effect on rats with MI/R injury when compared with the other pretreatment groups. Furthermore, the most effective duration of SM + DOO treatment was 30 min and the least effective duration was 180 min. Treatment with SM + DOO also significantly (P<0.01) reduced the number of terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling-positive cells, tumor necrosis factor-α andinterleukin-6 expression, and malondialdehyde content, and increased the serum and tissue activity of superoxide dismutase. These results indicated that the combined effects of SM + DOO may be more effective compared with the single pretreatments against MI/R injury in rats. This effect may be achieved partly through anti-apoptotic, antioxidant and anti-inflammatory activities. Therefore, SM + DOO may be considered an effective and promising novel strategy for the prophylaxis and treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Fei Mu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jialin Duan
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Haixu Bian
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ying Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yanrong Zhu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Guo Wei
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yue Guan
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yanhua Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Miaomiao Xi
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
28
|
Pan D, Li D. At the crossroads from bench to bedside: luteolin is a promising pharmacological agent against myocardial ischemia reperfusion injury. ANNALS OF TRANSLATIONAL MEDICINE 2017; 4:475. [PMID: 28090531 DOI: 10.21037/atm.2016.11.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Defeng Pan
- The First Clinical College, Nanjing Traditional Chinese Medicine University, Nanjing, China
| | - Dongye Li
- The First Clinical College, Nanjing Traditional Chinese Medicine University, Nanjing, China;; Institute of cardiovascular diseases, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
29
|
|
30
|
Maiese K. Targeting molecules to medicine with mTOR, autophagy and neurodegenerative disorders. Br J Clin Pharmacol 2016; 82:1245-1266. [PMID: 26469771 PMCID: PMC5061806 DOI: 10.1111/bcp.12804] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 10/11/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders are significantly increasing in incidence as the age of the global population continues to climb with improved life expectancy. At present, more than 30 million individuals throughout the world are impacted by acute and chronic neurodegenerative disorders with limited treatment strategies. The mechanistic target of rapamycin (mTOR), also known as the mammalian target of rapamycin, is a 289 kDa serine/threonine protein kinase that offers exciting possibilities for novel treatment strategies for a host of neurodegenerative diseases that include Alzheimer's disease, Parkinson's disease, Huntington's disease, epilepsy, stroke and trauma. mTOR governs the programmed cell death pathways of apoptosis and autophagy that can determine neuronal stem cell development, precursor cell differentiation, cell senescence, cell survival and ultimate cell fate. Coupled to the cellular biology of mTOR are a number of considerations for the development of novel treatments involving the fine control of mTOR signalling, tumourigenesis, complexity of the apoptosis and autophagy relationship, functional outcome in the nervous system, and the intimately linked pathways of growth factors, phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), AMP activated protein kinase (AMPK), silent mating type information regulation two homologue one (Saccharomyces cerevisiae) (SIRT1) and others. Effective clinical translation of the cellular signalling mechanisms of mTOR offers provocative avenues for new drug development in the nervous system tempered only by the need to elucidate further the intricacies of the mTOR pathway.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey, 07101, USA.
| |
Collapse
|
31
|
Biasutto L, Azzolini M, Szabò I, Zoratti M. The mitochondrial permeability transition pore in AD 2016: An update. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2515-30. [PMID: 26902508 DOI: 10.1016/j.bbamcr.2016.02.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Over the past 30years the mitochondrial permeability transition - the permeabilization of the inner mitochondrial membrane due to the opening of a wide pore - has progressed from being considered a curious artifact induced in isolated mitochondria by Ca(2+) and phosphate to a key cell-death-inducing process in several major pathologies. Its relevance is by now universally acknowledged and a pharmacology targeting the phenomenon is being developed. The molecular nature of the pore remains to this day uncertain, but progress has recently been made with the identification of the FOF1 ATP synthase as the probable proteic substrate. Researchers sharing this conviction are however divided into two camps: these believing that only the ATP synthase dimers or oligomers can form the pore, presumably in the contact region between monomers, and those who consider that the ring-forming c subunits in the FO sector actually constitute the walls of the pore. The latest development is the emergence of a new candidate: Spastic Paraplegia 7 (SPG7), a mitochondrial AAA-type membrane protease which forms a 6-stave barrel. This review summarizes recent developments of research on the pathophysiological relevance and on the molecular nature of the mitochondrial permeability transition pore. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Michele Azzolini
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Ildikò Szabò
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biology, Viale G. Colombo 3, 35121 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
32
|
Epps JA, Smart NA. Remote ischaemic conditioning in the context of type 2 diabetes and neuropathy: the case for repeat application as a novel therapy for lower extremity ulceration. Cardiovasc Diabetol 2016; 15:130. [PMID: 27613524 PMCID: PMC5018170 DOI: 10.1186/s12933-016-0444-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/19/2016] [Indexed: 02/07/2023] Open
Abstract
An emerging treatment modality for reducing damage caused by ischaemia–reperfusion injury is ischaemic conditioning. This technique induces short periods of ischaemia that have been found to protect against a more significant ischaemic insult. Remote ischaemic conditioning (RIC) can be administered more conveniently and safely, by inflation of a pneumatic blood pressure cuff to a suprasystolic pressure on a limb. Protection is then transferred to a remote organ via humoral and neural pathways. The diabetic state is particularly vulnerable to ischaemia–reperfusion injury, and ischaemia is a significant cause of many diabetic complications, including the diabetic foot. Despite this, studies utilising ischaemic conditioning and RIC in type 2 diabetes have often been disappointing. A newer strategy, repeat RIC, involves the repeated application of short periods of limb ischaemia over days or weeks. It has been demonstrated that this improves endothelial function, skin microcirculation, and modulates the systemic inflammatory response. Repeat RIC was recently shown to be beneficial for healing in lower extremity diabetic ulcers. This article summarises the mechanisms of RIC, and the impact that type 2 diabetes may have upon these, with the role of neural mechanisms in the context of diabetic neuropathy a focus. Repeat RIC may show more promise than RIC in type 2 diabetes, and its potential mechanisms and applications will also be explored. Considering the high costs, rates of chronicity and serious complications resulting from diabetic lower extremity ulceration, repeat RIC has the potential to be an effective novel advanced therapy for this condition.
Collapse
Affiliation(s)
- J A Epps
- School of Science and Technology, The University of New England, Armidale, NSW, 2351, Australia
| | - N A Smart
- School of Science and Technology, The University of New England, Armidale, NSW, 2351, Australia.
| |
Collapse
|
33
|
Ma L, Chuang CC, Weng W, Zhao L, Zheng Y, Zhang J, Zuo L. Paeonol Protects Rat Heart by Improving Regional Blood Perfusion during No-Reflow. Front Physiol 2016; 7:298. [PMID: 27493631 PMCID: PMC4954854 DOI: 10.3389/fphys.2016.00298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/28/2016] [Indexed: 01/22/2023] Open
Abstract
No-reflow phenomenon, defined as inadequate perfusion of myocardium without evident artery obstruction, occurs at a high incidence after coronary revascularization. The mechanisms underlying no-reflow is only partially understood. It is commonly caused by the swelling of endothelial cells, neutrophil accumulation, and vasoconstriction, which are all related to acute inflammation. Persistent no-reflow can lead to hospitalization and mortality. However, an effective preventive intervention has not yet been established. We have previously found that paeonol, an active extraction from the root of Paeonia suffruticosa, can benefit the heart function by inhibiting tissue damage after ischemia, reducing inflammation, and inducing vasodilatation. To further investigate the potential cardioprotective action of paeonol on no-reflow, healthy male Wistar rats were randomly divided into four groups: sham, ischemia-reperfusion (I/R) injury (left anterior descending coronary artery was ligated for 4 h followed by reperfusion for 8 h), and I/R injury pretreated with paeonol at two different doses. Real-time myocardial contrast echocardiography was used to monitor regional blood perfusion and cardiac functions. Our data indicated that paeonol treatment significantly reduces myocardial infarct area and no-reflow area (n = 8; p < 0.05). Regional myocardial perfusion (A·β) and cardiac functions such as ejection fraction, stroke volume, and fractional shortening were elevated by paeonol (n = 8; p < 0.05). Paeonol also lowered the serum levels of lactate dehydrogenase, creatine kinase, cardiac troponin T, and C-reactive protein, as indices of myocardial injury. Paeonol exerts beneficial effects on attenuating I/R-associated no-reflow injuries, and may be considered as a potential preventive treatment for cardiac diseases or post-coronary revascularization in which no-reflow often occurs.
Collapse
Affiliation(s)
- Lina Ma
- Graduate School, Beijing University of Chinese MedicineBeijing, China; Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical SciencesBeijing, China
| | - Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of MedicineColumbus, OH, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State UniversityColumbus, OH, USA
| | - Weiliang Weng
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Le Zhao
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Yongqiu Zheng
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Jinyan Zhang
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of MedicineColumbus, OH, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State UniversityColumbus, OH, USA
| |
Collapse
|
34
|
Ma LN, Li LD, Li SC, Hao XM, Zhang JY, He P, Li YK. Allicin improves cardiac function by protecting against apoptosis in rat model of myocardial infarction. Chin J Integr Med 2016; 23:589-597. [DOI: 10.1007/s11655-016-2523-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 12/28/2022]
|
35
|
Gonca E, Rapposelli S, Darıcı F, Digiacomo M, Yılmaz Z. Antiarrhythmic activity of a new spiro-cyclic benzopyran activator of the cardiac mitochondrial ATP dependent potassium channels. Arch Pharm Res 2016; 39:1212-22. [PMID: 27357534 DOI: 10.1007/s12272-016-0779-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/18/2016] [Indexed: 11/24/2022]
Abstract
'Compound A' (4(ı)-(N-(4-acetamidobenzyl))-2,2-dimethyl-2,3-dihydro-5(ı)H-spiro[chromene-4,2(ı)-[1,4]oxazinan]-5(ı)-one) is a new spiro-cyclic benzopyran activator of the mitochondrial ATP-dependent potassium channels (mitoKATP). We researched the effect of compound A on ischemia/reperfusion (I/R)-induced ventricular arrhythmias. We also tested the hypothesis that the application of the activation of mitoKATP in combination with the inhibition of sarcolemmal ATP-dependent potassium channels (sarcKATP) may produce a stronger antiarrhythmic effect. In anesthetized rats, myocardial ischemia was performed by ligating the left main coronary artery followed by reperfusion. At a dose of 10 mg/kg, compound A significantly decreased arrhythmia scores and the total length of arrhythmias, whereas this was found to be ineffective at a dose of 3 mg/kg. Pre-treatment with 5-HD, a selective mitoKATP blocker, abolished the antiarrhythmic effect of compound A. Both diazoxide, a selective mitoKATP opener and HMR 1098, a selective sarcKATP blocker, significantly decreased the total length of arrhythmias. However, the combination of neither diazoxide nor compound A with HMR 1098 showed no additional therapeutic benefit. These results reveal that compound A may have a dose-dependent antiarrythmic effect, which is more pronounced than the antiarrhythmic effect of diazoxide. The antiarrhythmic effect of compound A may possibly depend on mitoKATP activation.
Collapse
Affiliation(s)
- Ersöz Gonca
- Department of Biology, Faculty of Art and Sciences, Bülent Ecevit University, Zonguldak, Turkey.
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Faruk Darıcı
- Department of Biology, Faculty of Art and Sciences, Bülent Ecevit University, Zonguldak, Turkey
| | - Maria Digiacomo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Zehra Yılmaz
- Department of Pharmacology, Faculty of Medicine, Harran University, Şanliurfa, Turkey
| |
Collapse
|
36
|
Riquelme JA, Chavez MN, Mondaca-Ruff D, Bustamante M, Vicencio JM, Quest AFG, Lavandero S. Therapeutic targeting of autophagy in myocardial infarction and heart failure. Expert Rev Cardiovasc Ther 2016; 14:1007-19. [PMID: 27308848 DOI: 10.1080/14779072.2016.1202760] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Myocardial infarction (MI) is the leading cause of death. When MI is not lethal, heart failure (HF) is a major consequence with high prevalence and poor prognosis. The targeting of autophagy represents a potentially therapeutic approach for the treatment of both pathologies. AREAS COVERED PubMed searches were performed to discuss the current state of the art regarding the role of autophagy in MI and HF. We review available and potential approaches to modulate autophagy from a pharmacological and genetic perspective. We also discuss the targeting of autophagy in myocardial regeneration. Expert commentary: The targeting of autophagy has potential for the treatment of MI and HF. Autophagy is a process that takes place in virtually all cells of the body and thus, in order to evaluate this therapeutic approach in clinical trials, strategies that specifically target this process in the myocardium is required to avoid unwanted effects in other organs.
Collapse
Affiliation(s)
- Jaime A Riquelme
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Myra N Chavez
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,b FONDAP Center for Genome Regulation, Facultad de Ciencias , Universidad de Chile , Santiago , Chile
| | - David Mondaca-Ruff
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Mario Bustamante
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,c Advanced Center for Chronic Disease (ACCDiS), Division Enfermedades Cardiovasculares, Facultad de Medicina , Pontificia Universidad Catolica de Chile , Santiago , Chile
| | - Jose Miguel Vicencio
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,d Cancer Institute , University College London , London , UK
| | - Andrew F G Quest
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Sergio Lavandero
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,e Department of Internal Medicine, Cardiology Division , University of Texas Southwestern Medical Center , Dallas , TX , USA
| |
Collapse
|
37
|
Intracoronary administration of different doses of anisodamine in primary percutaneous coronary intervention. Coron Artery Dis 2016; 27:302-10. [DOI: 10.1097/mca.0000000000000366] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Vanezis AP, Rodrigo GC, Squire IB, Samani NJ. Remote ischaemic conditioning and remodelling following myocardial infarction: current evidence and future perspectives. Heart Fail Rev 2016; 21:635-43. [PMID: 27177446 PMCID: PMC4983281 DOI: 10.1007/s10741-016-9560-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Remote ischaemic conditioning (rIC) has demonstrated its effectiveness as a powerful cardioprotective tool in number of preclinical and limited clinical settings. More recently, ischaemic postconditioning given after an ischaemic event such as a myocardial infarction (MI) has shown not only to reduce infarct size but also to have beneficial effects on acute remodelling post-MI and to reduce the burden of heart failure and other detrimental outcomes. Building on this platform, repeated rIC over a number of days has the potential to augment the protective process even further. This review considers the current evidence base from which the concept of rIC in the setting of post-MI remodelling has grown. It also discusses the ongoing and planned clinical trials which are attempting to elucidate whether the protection imparted by rIC in the preclinical setting can be translated to the clinic and become a realistic weapon in the clinician’s armoury to tackle acute remodelling and heart failure post-MI.
Collapse
Affiliation(s)
- A P Vanezis
- Department of Cardiovascular Sciences, Glenfield General Hospital, University of Leicester, Leicester, LE3 9QP, UK.
| | - G C Rodrigo
- Department of Cardiovascular Sciences, Glenfield General Hospital, University of Leicester, Leicester, LE3 9QP, UK
| | - I B Squire
- Department of Cardiovascular Sciences, Glenfield General Hospital, University of Leicester, Leicester, LE3 9QP, UK
| | - N J Samani
- Department of Cardiovascular Sciences, Glenfield General Hospital, University of Leicester, Leicester, LE3 9QP, UK
| |
Collapse
|
39
|
Fukushima S, Miyagawa S, Sakai Y, Sawa Y. A sustained-release drug-delivery system of synthetic prostacyclin agonist, ONO-1301SR: a new reagent to enhance cardiac tissue salvage and/or regeneration in the damaged heart. Heart Fail Rev 2016; 20:401-13. [PMID: 25708182 PMCID: PMC4464640 DOI: 10.1007/s10741-015-9477-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac failure is a major cause of mortality and morbidity worldwide, since the standard treatment for cardiac failure in the clinical practice is chiefly to focus on removal of insults against the heart or minimisation of additional factors to exacerbate cardiac failure, but not on regeneration of the damaged cardiac tissue. A synthetic prostacyclin agonist, ONO-1301, has been developed as a long-acting drug for acute and chronic pathologies related to regional ischaemia, inflammation and/or interstitial fibrosis by pre-clinical studies. In addition, poly-lactic co-glycolic acid-polymerised form of ONO-1301, ONO-1301SR, was generated to achieve a further sustained release of this drug into the targeted region. This unique reagent has been shown to act on fibroblasts, vascular smooth muscle cells and endothelial cells in the tissue via the prostaglandin IP receptor to exert paracrinal release of multiple protective factors, such as hepatocyte growth factor, vascular endothelial growth factor or stromal cell-derived factor-1, into the adjacent damaged tissue, which is salvaged and/or regenerated as a result. Our laboratory developed a new surgical approach to treat acute and chronic cardiac failure using a variety of animal models, in which ONO-1301SR is directly placed over the cardiac surface to maximise the therapeutic effects and minimise the systemic complications. This review summarises basic and pre-clinical information of ONO-1301 and ONO-1301SR as a new reagent to enhance tissue salvage and/or regeneration, with a particular focus on the therapeutic effects on acute and chronic cardiac failure and underlying mechanisms, to explore a potential in launching the clinical study.
Collapse
Affiliation(s)
- Satsuki Fukushima
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan,
| | | | | | | |
Collapse
|
40
|
Hedegaard ER, Johnsen J, Povlsen JA, Jespersen NR, Shanmuganathan JA, Laursen MR, Kristiansen SB, Simonsen U, Botker HE. Inhibition of KV7 Channels Protects the Rat Heart against Myocardial Ischemia and Reperfusion Injury. ACTA ACUST UNITED AC 2016; 357:94-102. [DOI: 10.1124/jpet.115.230409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/05/2016] [Indexed: 02/06/2023]
|
41
|
Tritto I, Ambrosio G. Why does pre-clinical success in cardioprotection fail at the bedside? Cardiovasc Res 2016; 109:189-90. [PMID: 26705363 DOI: 10.1093/cvr/cvv277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Isabella Tritto
- Division of Cardiology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Giuseppe Ambrosio
- Division of Cardiology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
42
|
Luo SY, Chen S, Qin YD, Chen ZW. Urotensin-ⅡReceptor Antagonist SB-710411 Protects Rat Heart against Ischemia-Reperfusion Injury via RhoA/ROCK Pathway. PLoS One 2016; 11:e0146094. [PMID: 26771557 PMCID: PMC4714846 DOI: 10.1371/journal.pone.0146094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 12/14/2015] [Indexed: 12/18/2022] Open
Abstract
Aim SB-710411 is a rat selective urotensin-II (U-II) receptor antagonist, which can block U-II-induced contraction of the aorta and inhibit U-II-induced myocardial fibrosis in rats. However, the effect of SB-710411 on myocardial ischemia-reperfusion (I/R) injury is unclear. The present study was designed to investigate whether SB-710411 has a protective effect on myocardial I/R injury in rats and the possible mechanisms. Methods and Results Myocardial I/R injury was induced by occluding the left anterior descending coronary artery in adult male Sprague-Dawley rats. Hemodynamic parameters, electrocardiogram (ECG), infarct size, histological alteration, lactate dehydrogenase (LDH), creatine phosphokinase-MB (CK-MB), cardiac troponin I (cTnI), RhoA, and the protein expressions of U-II receptor (UTR), ROCK1 and ROCK2 were evaluated. Cardiac I/R injury significantly up-regulated the expressions of UTR, ROCK1 and ROCK2 proteins in rat myocardium. SB-710411 1.0 and 2.0 μg/kg significantly reduced cardiac I/R-induced the infarct size and histological damage in rat myocardium, markedly inhibited the changes of hemodynamic parameters and the increases of ST-segment in ECG, the serum LDH and CK-MB activities and cTnI level in rats subjected to myocardial I/R injury. Furthermore, SB-710411 obviously prevented myocardial I/R-increased RhoA activity and UTR, ROCK1 and ROCK2 protein expressions. Conclusions Our results indicate that cardiac I/R injury increases myocardial UTR expression, and SB-710411 has a potent protective effect on myocardial I/R injury in rats. The cardioprotection may be associated with the inhibition of UTR-RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Sheng-Yong Luo
- Department of Pharmacology, Anhui Medical University, Hefei, Anhui, China.,Anhui academy of medical sciences, Hefei, Anhui, China
| | - Shuo Chen
- Xinglin College of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yi-De Qin
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui, China
| | - Zhi-Wu Chen
- Department of Pharmacology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
43
|
Hu M, Ye P, Liao H, Chen M, Yang F. Metformin Protects H9C2 Cardiomyocytes from High-Glucose and Hypoxia/Reoxygenation Injury via Inhibition of Reactive Oxygen Species Generation and Inflammatory Responses: Role of AMPK and JNK. J Diabetes Res 2016; 2016:2961954. [PMID: 27294149 PMCID: PMC4884853 DOI: 10.1155/2016/2961954] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/08/2016] [Accepted: 03/29/2016] [Indexed: 12/20/2022] Open
Abstract
Metformin is a first-line drug for the management of type 2 diabetes. Recent studies suggested cardioprotective effects of metformin against ischemia/reperfusion injury. However, it remains elusive whether metformin provides direct protection against hypoxia/reoxygenation (H/R) injury in cardiomyocytes under normal or hyperglycemic conditions. This study in H9C2 rat cardiomyoblasts was designed to determine cell viability under H/R and high-glucose (HG, 33 mM) conditions and the effects of cotreatment with various concentrations of metformin (0, 1, 5, and 10 mM). We further elucidated molecular mechanisms underlying metformin-induced cytoprotection, especially the possible involvement of AMP-activated protein kinase (AMPK) and Jun NH(2)-terminal kinase (JNK). Results indicated that 5 mM metformin improved cell viability, mitochondrial integrity, and respiratory chain activity under HG and/or H/R (P < 0.05). The beneficial effects were associated with reduced levels of reactive oxygen species generation and proinflammatory cytokines (TNF-α, IL-1α, and IL-6) (P < 0.05). Metformin enhanced phosphorylation level of AMPK and suppressed HG + H/R induced JNK activation. Inhibitor of AMPK (compound C) or activator of JNK (anisomycin) abolished the cytoprotective effects of metformin. In conclusion, our study demonstrated for the first time that metformin possessed direct cytoprotective effects against HG and H/R injury in cardiac cells via signaling mechanisms involving activation of AMPK and concomitant inhibition of JNK.
Collapse
Affiliation(s)
- Mingyan Hu
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Ping Ye
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Hua Liao
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Manhua Chen
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Feiyan Yang
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
- *Feiyan Yang:
| |
Collapse
|
44
|
Molecular Characterization of Reactive Oxygen Species in Myocardial Ischemia-Reperfusion Injury. BIOMED RESEARCH INTERNATIONAL 2015; 2015:864946. [PMID: 26509170 PMCID: PMC4609796 DOI: 10.1155/2015/864946] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/11/2015] [Indexed: 12/11/2022]
Abstract
Myocardial ischemia-reperfusion (I/R) injury is experienced by individuals suffering from cardiovascular diseases such as coronary heart diseases and subsequently undergoing reperfusion treatments in order to manage the conditions. The occlusion of blood flow to the tissue, termed ischemia, can be especially detrimental to the heart due to its high energy demand. Several cellular alterations have been observed upon the onset of ischemia. The danger created by cardiac ischemia is somewhat paradoxical in that a return of blood to the tissue can result in further damage. Reactive oxygen species (ROS) have been studied intensively to reveal their role in myocardial I/R injury. Under normal conditions, ROS function as a mediator in many cell signaling pathways. However, stressful environments significantly induce the generation of ROS which causes the level to exceed body's antioxidant defense system. Such altered redox homeostasis is implicated in myocardial I/R injury. Despite the detrimental effects from ROS, low levels of ROS have been shown to exert a protective effect in the ischemic preconditioning. In this review, we will summarize the detrimental role of ROS in myocardial I/R injury, the protective mechanism induced by ROS, and potential treatments for ROS-related myocardial injury.
Collapse
|
45
|
Almohammedi A, Kapetanaki SM, Hudson AJ, Storey NM. Monitoring Changes in the Redox State of Myoglobin in Cardiomyocytes by Raman Spectroscopy Enables the Protective Effect of NO Donors to Be Evaluated. Anal Chem 2015; 87:10605-12. [PMID: 26407187 DOI: 10.1021/acs.analchem.5b03103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Raman microspectroscopy has been used to monitor changes in the redox and ligand-coordination states of the heme complex in myoglobin during the preconditioning of ex vivo cardiomyocytes with pharmacological drugs that release nitric oxide (NO). These chemical agents are known to confer protection on heart tissue against ischemia-reperfusion injury. Subsequent changes in the redox and ligand-coordination states during experimental simulations of ischemia and reperfusion have also been monitored. We found that these measurements, in real time, could be used to evaluate the preconditioning treatment of cardiomyocytes and to predict the likelihood of cell survival following a potentially lethal period of ischemia. Evaluation of the preconditioning treatment was done at the single-cell level. The binding of NO to myoglobin, giving a 6-coordinate ferrous-heme complex, was inferred from the measured Raman bands of a cardiomyocyte by comparison to pure solution of the protein in the presence of NO. A key change in the Raman spectrum was observed after perfusion of the NO-donor was completed, where, if the preconditioning treatment was successful, the bands corresponding to the nitrosyl complex were replaced by bands corresponding to metmyoglobin, Mb(III). An observation of Mb(III) bands in the Raman spectrum was made for all of the cardiomyocytes that recovered contractile function, whereas the absence of Mb(III) bands always indicated that the cardiomyocyte would be unable to recover contractile function following the simulated conditions of ischemia and reperfusion in these experiments.
Collapse
Affiliation(s)
| | | | | | - Nina M Storey
- Department of Cell Physiology and Pharmacology, University of Leicester , Leicester, LE1 9HN, United Kingdom
| |
Collapse
|
46
|
The cardioprotective effect of salidroside against myocardial ischemia reperfusion injury in rats by inhibiting apoptosis and inflammation. Apoptosis 2015; 20:1433-43. [DOI: 10.1007/s10495-015-1174-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Alánová P, Husková Z, Kopkan L, Sporková A, Jíchová Š, Neckář J, Imig JD, Klevstig M, Kolář F, Rami Reddy N, Falck JR, Sadowski J, Nishiyama A, Kramer HJ, Melenovský V, Červenková L, Kujal P, Vernerová Z, Červenka L. Orally active epoxyeicosatrienoic acid analog does not exhibit antihypertensive and reno- or cardioprotective actions in two-kidney, one-clip Goldblatt hypertensive rats. Vascul Pharmacol 2015; 73:45-56. [PMID: 26304700 DOI: 10.1016/j.vph.2015.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 07/20/2015] [Accepted: 08/20/2015] [Indexed: 12/20/2022]
Abstract
This study examined the effects of a novel orally active 14,15-epoxyeicosatrienoic acid analog (EET-A) on blood pressure (BP) and myocardial infarct size (IS) in two-kidney, one-clip (2K1C) Goldblatt hypertensive rats during sustained phase of hypertension. Between days 31 and 35 after clip placement the rats were treated with EET-A and BP was monitored by radiotelemetry; sham-operated normotensive rats were used as controls. Tissue concentrations of epoxyeicosatrienoic acids served as a marker of production of epoxygenase metabolites. The rats were subjected to acute myocardial ischemia/reperfusion (I/R) injury and IS was determined. We found that EET-A treatment did not lower BP in 2K1C rats and did not alter availability of biologically active epoxygenase metabolites in 2K1C or in sham-operated rats. The myocardial IS was significantly smaller in untreated 2K1C rats as compared with normotensive controls and EET-A reduced it in controls but not in 2K1C rats. Our findings suggest that during the phase of sustained hypertension 2K1C Goldblatt hypertensive rats exhibit increased cardiac tolerance to I/R injury as compared with normotensive controls, and that in this animal model of human renovascular hypertension short-term treatment with EET-A does not induce any antihypertensive and cardioprotective actions.
Collapse
Affiliation(s)
- Petra Alánová
- Department of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| | - Zuzana Husková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Libor Kopkan
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Alexandra Sporková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Šárka Jíchová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Jan Neckář
- Department of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic; Department of Pharmacology and Toxicology, Medical College of Wisconsin, WI, USA.
| | - John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, WI, USA.
| | - Martina Klevstig
- Department of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| | - František Kolář
- Department of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| | - N Rami Reddy
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Janusz Sadowski
- Department of Renal and Body Fluid Physiology, M. Mossakowski Medical Research Centre, Polish Academy of Science, Warsaw, Poland.
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kagawa, Japan.
| | - Herbert J Kramer
- Section of Nephrology, Medical Polyclinic, Department of Medicine, University of Bonn, Bonn, Germany.
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Lenka Červenková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Petr Kujal
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Pathology, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Zdenka Vernerová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Pathology, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Pathophysiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
48
|
Bromage DI, Yellon DM. The pleiotropic effects of metformin: time for prospective studies. Cardiovasc Diabetol 2015; 14:109. [PMID: 26271457 PMCID: PMC4536861 DOI: 10.1186/s12933-015-0273-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/03/2015] [Indexed: 12/19/2022] Open
Abstract
The global prevalence of diabetes has risen to epidemic proportions and the trend is predicted to continue. The consequent burden of cardiovascular morbidity and mortality is a major public health concern and new treatments are required to mitigate the deleterious effects of cardiovascular disease in diabetic patients. Ischaemia–reperfusion injury is well known to exacerbate the harmful effects of acute myocardial infarction and subsequent therapeutic reperfusion, and several mechanical and pharmacological approaches to mitigating this injury have been investigated. Metformin, which is cheap, relatively safe and widely used in type 2 diabetes, is one such pharmacotherapy with considerable pre-clinical evidence for cardioprotective utility beyond its glucose-lowering effect. However, despite convincing basic evidence its translation to clinical application has largely been limited to studies of cardiovascular risk. There are several barriers to prospective randomized assessment in the context of acute myocardial infarction, not least the accessibility and already widespread use of metformin among patients with type 2 diabetes at high risk of cardiovascular events. In the place of class 1 evidence, well-designed prospective cohort studies of the potential pleiotropic utility of metformin in cardiovascular disease, and particularly its benefit in ischaemia–reperfusion injury, are needed. Given the availability of metformin worldwide, this is particularly true in low- and middle-income countries where the optimal therapy for acute myocardial infarction, primary percutaneous coronary intervention, may not be available, and instead patients are managed with thrombolysis. As this is less effective, metformin as an adjunct to thrombolysis (or PPCI) could represent an effective, cheap means of cardioprotection with global relevance.
Collapse
Affiliation(s)
- Daniel I Bromage
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
49
|
Jiang H, Xiao J, Kang B, Zhu X, Xin N, Wang Z. PI3K/SGK1/GSK3β signaling pathway is involved in inhibition of autophagy in neonatal rat cardiomyocytes exposed to hypoxia/reoxygenation by hydrogen sulfide. Exp Cell Res 2015; 345:134-40. [PMID: 26163895 DOI: 10.1016/j.yexcr.2015.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/19/2015] [Accepted: 07/06/2015] [Indexed: 11/16/2022]
Abstract
Excessive autophagy aggravates myocardial ischemia/reperfusion (IR) injury. Hydrogen sulfide (H2S) has been shown to possess a strong cardioprotective effect due to its anti-necrosis, anti-apoptosis, anti-oxidant and anti-inflammatory properties. Our previous study showed that H2S could also protect the myocardium against IR injury through its anti-autophagy effect in vivo, but the underlying mechanism remains unclear. The aim of the present study was to determine whether PI3K/SGK1/GSK3β signaling pathway was involved in the anti-autophagy effect of H2S against myocardial hypoxia/reoxygenation (HR) injury in vitro. Autophagy was significantly increased in cardiomyocytes subjected to HR, but it was down-regulated by H2S (NaHS donor). Blocking PI3K by LY294002 (a PI3K inhibitor) or knocking down SGK1 by SGK1 siRNA augmented autophagy and attenuated the anti-autophagy effect of H2S. However, blocking GSK3β by tws119 (a GSK3β inhibitor) produced an opposite effect. In addition, while treatment of neonatal rat cardiomyocytes with HR reduced cell viability and augmented cell injury, H2S significantly reversed it. Blocking PI3K or knocking down SGK1 aggravated HR injury and weakened the protective effect of H2S, while blocking GSK3β produced an opposite effect. In conclusion, H2S can inhibit autophagy in neonatal rat cardiomyocytes exposed to H/R and exert a cardioprotective effect at least partly by regulating PI3K/SGK1/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Huan Jiang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bo Kang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Ni Xin
- Department of Physiology, Second Military Medical University, Shanghai, China.
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
50
|
Lee S, Hutchinson M, Staikopoulos V, Saint D. Amitriptyline pharmacologically preconditions rat hearts against cardiac ischemic–reperfusion injury. Int J Cardiol 2015; 190:353-9. [DOI: 10.1016/j.ijcard.2015.04.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/16/2015] [Accepted: 04/03/2015] [Indexed: 11/16/2022]
|