1
|
Hou S, Yan X, Gao X, Jockusch S, Gibson KM, Shan Z, Bi L. Enhancing Cardiomyocyte Resilience to Ischemia-Reperfusion Injury: The Therapeutic Potential of an Indole-Peptide-Tempo Conjugate (IPTC). ACS OMEGA 2024; 9:39401-39418. [PMID: 39346824 PMCID: PMC11425819 DOI: 10.1021/acsomega.4c02725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 10/01/2024]
Abstract
Ischemia/reperfusion (I/R) injury leads to apoptosis and extensive cellular and mitochondrial damage, triggered by the early generation and subsequent accumulation of mitochondrial reactive oxygen species (mtROS). This condition not only contributes to the pathology of I/R injury itself but is also implicated in a variety of other diseases, especially within the cardiovascular domain. Addressing mitochondrial oxidative stress thus emerges as a critical therapeutic target. In this context, our study introduces an indole-peptide-tempo conjugate (IPTC), a compound designed with dual functionalities: antioxidative properties and the ability to modulate autophagy. Our findings reveal that IPTC effectively shields H9C2 cardiomyocytes against hypoxia/reoxygenation (H/R) damage, primarily through counteracting mtROS overproduction linked to impaired mitophagy and mitochondrial dysfunction. We propose that IPTC operates by simultaneously reducing mtROS levels and inducing mitophagy, highlighting its potential as a novel therapeutic strategy for mitigating mitochondrial oxidative damage and, by extension, easing I/R injury and potentially other related cardiovascular conditions.
Collapse
Affiliation(s)
- Shanshan Hou
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Xin Yan
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Xiang Gao
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Steffen Jockusch
- Center of Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, United States
| | - K Michael Gibson
- Department of Pharmacotherapy, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Lanrong Bi
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
2
|
Chiari P, Fellahi JL. Myocardial protection in cardiac surgery: a comprehensive review of current therapies and future cardioprotective strategies. Front Med (Lausanne) 2024; 11:1424188. [PMID: 38962735 PMCID: PMC11220133 DOI: 10.3389/fmed.2024.1424188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 05/23/2024] [Indexed: 07/05/2024] Open
Abstract
Cardiac surgery with cardiopulmonary bypass results in global myocardial ischemia-reperfusion injury, leading to significant postoperative morbidity and mortality. Although cardioplegia is the cornerstone of intraoperative cardioprotection, a number of additional strategies have been identified. The concept of preconditioning and postconditioning, despite its limited direct clinical application, provided an essential contribution to the understanding of myocardial injury and organ protection. Therefore, physicians can use different tools to limit perioperative myocardial injury. These include the choice of anesthetic agents, remote ischemic preconditioning, tight glycemic control, optimization of respiratory parameters during the aortic unclamping phase to limit reperfusion injury, appropriate choice of monitoring to optimize hemodynamic parameters and limit perioperative use of catecholamines, and early reintroduction of cardioprotective agents in the postoperative period. Appropriate management before, during, and after cardiopulmonary bypass will help to decrease myocardial damage. This review aimed to highlight the current advancements in cardioprotection and their potential applications during cardiac surgery.
Collapse
Affiliation(s)
- Pascal Chiari
- Service d’Anesthésie Réanimation, Hôpital Universitaire Louis Pradel, Hospices Civils de Lyon, Lyon, France
- Laboratoire CarMeN, Inserm UMR 1060, Université Claude Bernard Lyon 1, Lyon, France
| | - Jean-Luc Fellahi
- Service d’Anesthésie Réanimation, Hôpital Universitaire Louis Pradel, Hospices Civils de Lyon, Lyon, France
- Laboratoire CarMeN, Inserm UMR 1060, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
3
|
Ravingerova T, Adameova A, Lonek L, Farkasova V, Ferko M, Andelova N, Kura B, Slezak J, Galatou E, Lazou A, Zohdi V, Dhalla NS. Is Intrinsic Cardioprotection a Laboratory Phenomenon or a Clinically Relevant Tool to Salvage the Failing Heart? Int J Mol Sci 2023; 24:16497. [PMID: 38003687 PMCID: PMC10671596 DOI: 10.3390/ijms242216497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases, especially ischemic heart disease, as a leading cause of heart failure (HF) and mortality, will not reduce over the coming decades despite the progress in pharmacotherapy, interventional cardiology, and surgery. Although patients surviving acute myocardial infarction live longer, alteration of heart function will later lead to HF. Its rising incidence represents a danger, especially among the elderly, with data showing more unfavorable results among females than among males. Experiments revealed an infarct-sparing effect of ischemic "preconditioning" (IPC) as the most robust form of innate cardioprotection based on the heart's adaptation to moderate stress, increasing its resistance to severe insults. However, translation to clinical practice is limited by technical requirements and limited time. Novel forms of adaptive interventions, such as "remote" IPC, have already been applied in patients, albeit with different effectiveness. Cardiac ischemic tolerance can also be increased by other noninvasive approaches, such as adaptation to hypoxia- or exercise-induced preconditioning. Although their molecular mechanisms are not yet fully understood, some noninvasive modalities appear to be promising novel strategies for fighting HF through targeting its numerous mechanisms. In this review, we will discuss the molecular mechanisms of heart injury and repair, as well as interventions that have potential to be used in the treatment of patients.
Collapse
Affiliation(s)
- Tanya Ravingerova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Adriana Adameova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 10 Odbojárov St., 832 32 Bratislava, Slovakia
| | - Lubomir Lonek
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Veronika Farkasova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Miroslav Ferko
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Natalia Andelova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Branislav Kura
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Jan Slezak
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Eleftheria Galatou
- School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (E.G.); (A.L.)
- Department of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (E.G.); (A.L.)
| | - Vladislava Zohdi
- Department of Anatomy, Faculty of Medicine, Comenius University in Bratislava, 24 Špitalska, 813 72 Bratislava, Slovakia;
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, 19 Innovation Walk, Clayton, VIC 3800, Australia
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada;
| |
Collapse
|
4
|
Osorio-Llanes E, Villamizar-Villamizar W, Ospino Guerra MC, Díaz-Ariza LA, Castiblanco-Arroyave SC, Medrano L, Mengual D, Belón R, Castellar-López J, Sepúlveda Y, Vásquez-Trincado C, Chang AY, Bolívar S, Mendoza-Torres E. Effects of Metformin on Ischemia/Reperfusion Injury: New Evidence and Mechanisms. Pharmaceuticals (Basel) 2023; 16:1121. [PMID: 37631036 PMCID: PMC10459572 DOI: 10.3390/ph16081121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The search for new drugs with the potential to ensure therapeutic success in the treatment of cardiovascular diseases has become an essential pathway to follow for health organizations and committees around the world. In June 2021, the World Health Organization listed cardiovascular diseases as one of the main causes of death worldwide, representing 32% of them. The most common is coronary artery disease, which causes the death of cardiomyocytes, the cells responsible for cardiac contractility, through ischemia and subsequent reperfusion, which leads to heart failure in the medium and short term. Metformin is one of the most-used drugs for the control of diabetes, which has shown effects beyond the control of hyperglycemia. Some of these effects are mediated by the regulation of cellular energy metabolism, inhibiting apoptosis, reduction of cell death through regulation of autophagy and reduction of mitochondrial dysfunction with further reduction of oxidative stress. This suggests that metformin may attenuate left ventricular dysfunction induced by myocardial ischemia; preclinical and clinical trials have shown promising results, particularly in the setting of acute myocardial infarction. This is a review of the molecular and pharmacological mechanisms of the cardioprotective effects of metformin during myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Estefanie Osorio-Llanes
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
- Allied Research Society S.A.S., Barranquilla 080001, Colombia;
- Global Disease Research Colombia, Barranquilla 080001, Colombia
| | - Wendy Villamizar-Villamizar
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - María Clara Ospino Guerra
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Luis Antonio Díaz-Ariza
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Sara Camila Castiblanco-Arroyave
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Luz Medrano
- Healthcare Pharmacy and Pharmacology Research Group, Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla 081007, Colombia; (L.M.); (D.M.); (S.B.)
| | - Daniela Mengual
- Healthcare Pharmacy and Pharmacology Research Group, Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla 081007, Colombia; (L.M.); (D.M.); (S.B.)
| | - Ricardo Belón
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Jairo Castellar-López
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Yanireth Sepúlveda
- Allied Research Society S.A.S., Barranquilla 080001, Colombia;
- Global Disease Research Colombia, Barranquilla 080001, Colombia
| | - César Vásquez-Trincado
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago 8370134, Chile;
| | - Aileen Y. Chang
- Department of Medicine, Faculty of Medicine, Foggy Bottom Campus, George Washington University, Washington, DC 20052, USA;
| | - Samir Bolívar
- Healthcare Pharmacy and Pharmacology Research Group, Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla 081007, Colombia; (L.M.); (D.M.); (S.B.)
| | - Evelyn Mendoza-Torres
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| |
Collapse
|
5
|
Huang Y, Zhou B. Mitochondrial Dysfunction in Cardiac Diseases and Therapeutic Strategies. Biomedicines 2023; 11:biomedicines11051500. [PMID: 37239170 DOI: 10.3390/biomedicines11051500] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondria are the main site of intracellular synthesis of ATP, which provides energy for various physiological activities of the cell. Cardiomyocytes have a high density of mitochondria and mitochondrial damage is present in a variety of cardiovascular diseases. In this paper, we describe mitochondrial damage in mitochondrial cardiomyopathy, congenital heart disease, coronary heart disease, myocardial ischemia-reperfusion injury, heart failure, and drug-induced cardiotoxicity, in the context of the key roles of mitochondria in cardiac development and homeostasis. Finally, we discuss the main current therapeutic strategies aimed at alleviating mitochondrial impairment-related cardiac dysfunction, including pharmacological strategies, gene therapy, mitochondrial replacement therapy, and mitochondrial transplantation. It is hoped that this will provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| |
Collapse
|
6
|
Ajzashokouhi AH, Rezaee R, Omidkhoda N, Karimi G. Natural compounds regulate the PI3K/Akt/GSK3β pathway in myocardial ischemia-reperfusion injury. Cell Cycle 2023; 22:741-757. [PMID: 36593695 PMCID: PMC10026916 DOI: 10.1080/15384101.2022.2161959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023] Open
Abstract
The PI3K/Akt/GSK3β pathway is crucial in regulating cardiomyocyte growth and survival. It has been shown that activation of this pathway alleviates the negative impact of ischemia-reperfusion. Glycogen synthase kinase-3 (GSK3β) induces apoptosis through stimulation of transcription factors, and its phosphorylation has been suggested as a new therapeutic target for myocardial ischemia-reperfusion injury (MIRI). GSK3β regulatory role is mediated by the reperfusion injury salvage kinase (RISK) pathway, and its inhibition by Akt activation blocks mitochondrial permeability transition pore (mPTP) opening and enhances myocardial survival. The present article discusses the involvement of the PI3K/Akt/GSK3β pathway in cardioprotective effects of natural products against MIRI.Abbreviations: Akt: protein kinase B; AMPK: AMP-activated protein kinase; ATP: adenosine triphosphate; Bad: bcl2-associated agonist of cell death; Bax: bcl2-associated x protein; Bcl-2: B-cell lymphoma 2; CK-MB: Creatine kinase-MB; CRP: C-reactive-protein; cTnI: cardiac troponin I; EGCG: Epigallocatechin-3-gallate; Enos: endothelial nitric oxide synthase; ER: endoplasmic reticulum; ERK ½: extracellular signal‑regulated protein kinase ½; GSK3β: glycogen synthase kinase-3; GSRd: Ginsenoside Rd; GSH: glutathione; GSSG: glutathione disulfide; HO-1: heme oxygenase-1; HR: hypoxia/reoxygenation; HSYA: Hydroxysafflor Yellow A; ICAM-1: Intercellular Adhesion Molecule 1; IKK-b: IκB kinase; IL: interleukin; IPoC: Ischemic postconditioning; IRI: ischemia-reperfusion injury; JNK: c-Jun N-terminal kinase; Keap1: kelch-like ECH-associated protein- 1; LDH: lactate dehydrogenase; LVEDP: left ventricular end diastolic pressure; LVP: left ventricle pressure; LVSP: left ventricular systolic pressure; MAPK: mitogen-activated protein kinase; MDA: malondialdehyde; MIRI: myocardial ischemia-reperfusion injury; MnSOD: manganese superoxide dismutase; mPTP: mitochondrial permeability transition pore; mtHKII: mitochondria-bound hexokinase II; Nrf-1: nuclear respiratory factor 1; Nrf2: nuclear factor erythroid 2-related factor; NO: nitric oxide; PGC-1α: peroxisome proliferator‑activated receptor γ coactivator‑1α; PI3K: phosphoinositide 3-kinases; RISK: reperfusion injury salvage kinase; ROS: reactive oxygen species; RSV: Resveratrol; SOD: superoxide dismutase; TFAM: transcription factor A mitochondrial; TNF-α: tumor necrosis factor-alpha; VEGF-B: vascular endothelial growth factor B.
Collapse
Affiliation(s)
| | - Ramin Rezaee
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Navid Omidkhoda
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Li A, Gao M, Liu B, Qin Y, Chen L, Liu H, Wu H, Gong G. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis 2022; 13:444. [PMID: 35534453 PMCID: PMC9085840 DOI: 10.1038/s41419-022-04906-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria are highly dynamic organelles that participate in ATP generation and involve calcium homeostasis, oxidative stress response, and apoptosis. Dysfunctional or damaged mitochondria could cause serious consequences even lead to cell death. Therefore, maintaining the homeostasis of mitochondria is critical for cellular functions. Mitophagy is a process of selectively degrading damaged mitochondria under mitochondrial toxicity conditions, which plays an essential role in mitochondrial quality control. The abnormal mitophagy that aggravates mitochondrial dysfunction is closely related to the pathogenesis of many diseases. As the myocardium is a highly oxidative metabolic tissue, mitochondria play a central role in maintaining optimal performance of the heart. Dysfunctional mitochondria accumulation is involved in the pathophysiology of cardiovascular diseases, such as myocardial infarction, cardiomyopathy and heart failure. This review discusses the most recent progress on mitophagy and its role in cardiovascular disease.
Collapse
Affiliation(s)
- Anqi Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Meng Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan Qin
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Lei Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hanyu Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Huayan Wu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
8
|
Li W, Xiang Z, Xing Y, Li S, Shi S. Mitochondria bridge HIF signaling and ferroptosis blockage in acute kidney injury. Cell Death Dis 2022; 13:308. [PMID: 35387983 PMCID: PMC8986825 DOI: 10.1038/s41419-022-04770-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 11/09/2022]
Abstract
AbstractFerroptosis, a form of regulated cell death, plays an important role in acute kidney injury (AKI). Previous studies have shown that prolyl hydroxylase domain protein (PHD) inhibitors that activate HIF signaling provide strong protection against AKI, which is characterized by marked cell death. However, the relationship between PHD inhibition/HIF signaling and ferroptosis in AKI has not been elucidated. Here, we review recent studies to explore the issue. First, we will review the literature concerning the functions of HIF in promoting mitophagy, suppressing mitochondrial respiration and modulating redox homeostasis. Second, we will describe the current understanding of ferroptosis and its role in AKI, particularly from the perspective of mitochondrial dysfunction. Finally, we will discuss the possibility that mitochondria link PHD inhibition/HIF signaling and ferroptosis in AKI. In conclusion, we propose that HIF may protect renal cells against ferroptosis in AKI by reducing mitochondrial oxidative stress and damage.
Collapse
|
9
|
Lewis MJ, Khaliulin I, Hall K, Suleiman MS. Cardioprotection of Immature Heart by Simultaneous Activation of PKA and Epac: A Role for the Mitochondrial Permeability Transition Pore. Int J Mol Sci 2022; 23:ijms23031720. [PMID: 35163640 PMCID: PMC8836102 DOI: 10.3390/ijms23031720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic and ionic changes during ischaemia predispose the heart to the damaging effects of reperfusion. Such changes and the resulting injury differ between immature and adult hearts. Therefore, cardioprotective strategies for adults must be tested in immature hearts. We have recently shown that the simultaneous activation of protein kinase A (PKA) and exchange protein activated by cAMP (Epac) confers marked cardioprotection in adult hearts. The aim of this study is to investigate the efficacy of this intervention in immature hearts and determine whether the mitochondrial permeability transition pore (MPTP) is involved. Isolated perfused Langendorff hearts from both adult and immature rats were exposed to global ischaemia and reperfusion injury (I/R) following control perfusion or perfusion after an equilibration period with activators of PKA and/or Epac. Functional outcome and reperfusion injury were measured and in parallel, mitochondria were isolated following 5 min of reperfusion to determine whether cardioprotective interventions involved changes in MPTP opening behaviour. Perfusion for 5 min preceding ischaemia of injury-matched adult and immature hearts with 5 µM 8-Br (8-Br-cAMP-AM), an activator of both PKA and Epac, led to significant reduction in post-reperfusion CK release and infarct size. Perfusion with this agent also led to a reduction in MPTP opening propensity in both adult and immature hearts. These data show that immature hearts are innately more resistant to I/R injury than adults, and that this is due to a reduced tendency of MPTP opening following reperfusion. Furthermore, simultaneous stimulation of PKA and Epac causes cardioprotection, which is additive to the innate resistance.
Collapse
Affiliation(s)
- Martin John Lewis
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
- Correspondence:
| | - Igor Khaliulin
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel;
| | - Katie Hall
- Bristol Medical School, University of Bristol, Bristol BS8 1TH, UK; (K.H.); (M.S.S.)
| | - M. Saadeh Suleiman
- Bristol Medical School, University of Bristol, Bristol BS8 1TH, UK; (K.H.); (M.S.S.)
| |
Collapse
|
10
|
Acetylcholine exerts cytoprotection against hypoxia/reoxygenation-induced apoptosis, autophagy and mitochondrial impairment through both muscarinic and nicotinic receptors. Apoptosis 2022; 27:233-245. [DOI: 10.1007/s10495-022-01715-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/25/2022]
|
11
|
Heart Failure after Cardiac Surgery: The Role of Halogenated Agents, Myocardial Conditioning and Oxidative Stress. Int J Mol Sci 2022; 23:ijms23031360. [PMID: 35163284 PMCID: PMC8836224 DOI: 10.3390/ijms23031360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/07/2022] Open
Abstract
Heart disease requires a surgical approach sometimes. Cardiac-surgery patients develop heart failure associated with ischemia induced during extracorporeal circulation. This complication could be decreased with anesthetic drugs. The cardioprotective effects of halogenated agents are based on pre- and postconditioning (sevoflurane, desflurane, or isoflurane) compared to intravenous hypnotics (propofol). We tried to put light on the shadows walking through the line of the halogenated anesthetic drugs’ effects in several enzymatic routes and oxidative stress, waiting for the final results of the ACDHUVV-16 clinical trial regarding the genetic modulation of this kind of drugs.
Collapse
|
12
|
Bland AR, Payne FM, Ashton JC, Jamialahmadi T, Sahebkar A. The cardioprotective actions of statins in targeting mitochondrial dysfunction associated with myocardial ischaemia-reperfusion injury. Pharmacol Res 2021; 175:105986. [PMID: 34800627 DOI: 10.1016/j.phrs.2021.105986] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/24/2022]
Abstract
During cardiac reperfusion after myocardial infarction, the heart is subjected to cascading cycles of ischaemia reperfusion injury (IRI). Patients presenting with this injury succumb to myocardial dysfunction resulting in myocardial cell death, which contributes to morbidity and mortality. New targeted therapies are required if the myocardium is to be protected from this injury and improve patient outcomes. Extensive research into the role of mitochondria during ischaemia and reperfusion has unveiled one of the most important sites contributing towards this injury; specifically, the opening of the mitochondrial permeability transition pore. The opening of this pore occurs during reperfusion and results in mitochondria swelling and dysfunction, promoting apoptotic cell death. Activation of mitochondrial ATP-sensitive potassium channels (mitoKATP) channels, uncoupling proteins, and inhibition of glycogen synthase kinase-3β (GSK3β) phosphorylation have been identified to delay mitochondrial permeability transition pore opening and reduce reactive oxygen species formation, thereby decreasing infarct size. Statins have recently been identified to provide a direct cardioprotective effect on these specific mitochondrial components, all of which reduce the severity of myocardial IRI, promoting the ability of statins to be a considerate preconditioning agent. This review will outline what has currently been shown in regard to statins cardioprotective effects on mitochondria during myocardial IRI.
Collapse
Affiliation(s)
- Abigail R Bland
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Fergus M Payne
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - John C Ashton
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Comità S, Femmino S, Thairi C, Alloatti G, Boengler K, Pagliaro P, Penna C. Regulation of STAT3 and its role in cardioprotection by conditioning: focus on non-genomic roles targeting mitochondrial function. Basic Res Cardiol 2021; 116:56. [PMID: 34642818 PMCID: PMC8510947 DOI: 10.1007/s00395-021-00898-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022]
Abstract
Ischemia–reperfusion injury (IRI) is one of the biggest challenges for cardiovascular researchers given the huge death toll caused by myocardial ischemic disease. Cardioprotective conditioning strategies, namely pre- and post-conditioning maneuvers, represent the most important strategies for stimulating pro-survival pathways essential to preserve cardiac health. Conditioning maneuvers have proved to be fundamental for the knowledge of the molecular basis of both IRI and cardioprotection. Among this evidence, the importance of signal transducer and activator of transcription 3 (STAT3) emerged. STAT3 is not only a transcription factor but also exhibits non-genomic pro-survival functions preserving mitochondrial function from IRI. Indeed, STAT3 is emerging as an influencer of mitochondrial function to explain the cardioprotection phenomena. Studying cardioprotection, STAT3 proved to be crucial as an element of the survivor activating factor enhancement (SAFE) pathway, which converges on mitochondria and influences their function by cross-talking with other cardioprotective pathways. Clearly there are still some functional properties of STAT3 to be discovered. Therefore, in this review, we highlight the evidence that places STAT3 as a promoter of the metabolic network. In particular, we focus on the possible interactions of STAT3 with processes aimed at maintaining mitochondrial functions, including the regulation of the electron transport chain, the production of reactive oxygen species, the homeostasis of Ca2+ and the inhibition of opening of mitochondrial permeability transition pore. Then we consider the role of STAT3 and the parallels between STA3/STAT5 in cardioprotection by conditioning, giving emphasis to the human heart and confounders.
Collapse
Affiliation(s)
- Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | - Saveria Femmino
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Cecilia Thairi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | | | - Kerstin Boengler
- Institute of Physiology, University of Giessen, Giessen, Germany
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| |
Collapse
|
14
|
Gurevich KG, Urakov AL, Fisher EL, Abzalilov TA, Khairzamanova KA, Yagudin TA, Samorodov AV. Possibilities of pharmacological correction of reperfusion injury of ischemic myocardium (review). REVIEWS ON CLINICAL PHARMACOLOGY AND DRUG THERAPY 2021; 19:259-267. [DOI: 10.17816/rcf193259-267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Timely and effective reperfusion in ischemia and reoxygenation in hypoxia of the heart muscle prevent myocardial infarction. Delayed reperfusion and reoxygenation in myocardial ischemia and hypoxia can cause reversible damage in it, which, with a favorable outcome, disappear without a trace. Excessively late reperfusion and reoxygenation inevitably ends with irreversible damage to the myocardium, which is widely known as a myocardial infarction, and which, together with other complications of cardiac ischemia, can cause disability and death of the patient. In recent years, reperfusion injury of the ischemic heart muscle has been recognized as an independent link in the pathogenesis of myocardial infarction. The mechanisms of this link of pathogenesis have been partially studied in experimental conditions. The phenomena of preconditioning and post-conditioning have been discovered, the effects of which are currently determined fairly reliably. After determining the mechanisms of reperfusion injury of the ischemic myocardium, the search and development of pharmacological agents capable of inducing such a phenomenon as cardioprotection began. In parallel, studies of specific microRNAs that claim to be diagnostic markers are being conducted, as well as the search for drugs that affect the level of their expression is being conducted. The information about the achieved successes in this direction is given.
Collapse
|
15
|
Dynamic Regulation of Cysteine Oxidation and Phosphorylation in Myocardial Ischemia-Reperfusion Injury. Cells 2021; 10:cells10092388. [PMID: 34572037 PMCID: PMC8469016 DOI: 10.3390/cells10092388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/02/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury significantly alters heart function following infarct and increases the risk of heart failure. Many studies have sought to preserve irreplaceable myocardium, termed cardioprotection, but few, if any, treatments have yielded a substantial reduction in clinical I/R injury. More research is needed to fully understand the molecular pathways that govern cardioprotection. Redox mechanisms, specifically cysteine oxidations, are acute and key regulators of molecular signaling cascades mediated by kinases. Here, we review the role of reactive oxygen species in modifying cysteine residues and how these modifications affect kinase function to impact cardioprotection. This exciting area of research may provide novel insight into mechanisms and likely lead to new treatments for I/R injury.
Collapse
|
16
|
Role of Oxidative Stress in Reperfusion following Myocardial Ischemia and Its Treatments. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6614009. [PMID: 34055195 PMCID: PMC8149218 DOI: 10.1155/2021/6614009] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/21/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022]
Abstract
Myocardial ischemia is a disease with high morbidity and mortality, for which reperfusion is currently the standard intervention. However, the reperfusion may lead to further myocardial damage, known as myocardial ischemia/reperfusion injury (MI/RI). Oxidative stress is one of the most important pathological mechanisms in reperfusion injury, which causes apoptosis, autophagy, inflammation, and some other damage in cardiomyocytes through multiple pathways, thus causing irreversible cardiomyocyte damage and cardiac dysfunction. This article reviews the pathological mechanisms of oxidative stress involved in reperfusion injury and the interventions for different pathways and targets, so as to form systematic treatments for oxidative stress-induced myocardial reperfusion injury and make up for the lack of monotherapy.
Collapse
|
17
|
Tsoumani M, Georgoulis A, Nikolaou PE, Kostopoulos IV, Dermintzoglou T, Papatheodorou I, Zoga A, Efentakis P, Konstantinou M, Gikas E, Kostomitsopoulos N, Papapetropoulos A, Lazou A, Skaltsounis AL, Hausenloy DJ, Tsitsilonis O, Tseti I, Di Lisa F, Iliodromitis EK, Andreadou I. Acute administration of the olive constituent, oleuropein, combined with ischemic postconditioning increases myocardial protection by modulating oxidative defense. Free Radic Biol Med 2021; 166:18-32. [PMID: 33582227 DOI: 10.1016/j.freeradbiomed.2021.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 02/06/2021] [Indexed: 12/13/2022]
Abstract
Oleuropein, one of the main polyphenolic constituents of olive, is cardioprotective against ischemia reperfusion injury (IRI). We aimed to assess the cardioprotection afforded by acute administration of oleuropein and to evaluate the underlying mechanism. Importantly, since antioxidant therapies have yielded inconclusive results in attenuating IRI-induced damage on top of conditioning strategies, we investigated whether oleuropein could enhance or imbed the cardioprotective manifestation of ischemic postconditioning (PostC). Oleuropein, given during ischemia as a single intravenous bolus dose reduced the infarct size compared to the control group both in rabbits and mice subjected to myocardial IRI. None of the inhibitors of the cardioprotective pathways, l-NAME, wortmannin and AG490, influence its infarct size limiting effects. Combined oleuropein and PostC cause further limitation of infarct size in comparison with PostC alone in both animal models. Oleuropein did not inhibit the calcium induced mitochondrial permeability transition pore opening in isolated mitochondria and did not increase cGMP production. To provide further insights to the different cardioprotective mechanism of oleuropein, we sought to characterize its anti-inflammatory potential in vivo. Oleuropein, PostC and their combination reduce inflammatory monocytes infiltration into the heart and the circulating monocyte cell population. Oleuropein's mechanism of action involves a direct protective effect on cardiomyocytes since it significantly increased their viability following simulated IRI as compared to non-treated cells. Οleuropein confers additive cardioprotection on top of PostC, via increasing the expression of the transcription factor Nrf-2 and its downstream targets in vivo. In conclusion, acute oleuropein administration during ischemia in combination with PostC provides robust and synergistic cardioprotection in experimental models of IRI by inducing antioxidant defense genes through Nrf-2 axis and independently of the classic cardioprotective signaling pathways (RISK, cGMP/PKG, SAFE).
Collapse
Affiliation(s)
- Maria Tsoumani
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Anastasios Georgoulis
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Panagiota-Efstathia Nikolaou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Ioannis V Kostopoulos
- Department of Animal and Human Physiology, Faculty of Biology, National and Kapodistrian University of Athens, 15784, Athens, Greece
| | - Theano Dermintzoglou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Ioanna Papatheodorou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Anastasia Zoga
- 2nd Department of Cardiology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Maria Konstantinou
- Laboratory of Pharmaceutical Analysis, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Evangelos Gikas
- Laboratory of Analytical Chemistry, Department of Chemistry, School of Science, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Centre of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527, Αthens Greece
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece; Centre of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527, Αthens Greece
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Alexios-Leandros Skaltsounis
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; The Hatter Cardiovascular Institute, University College London, London, UK; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan
| | - Ourania Tsitsilonis
- Department of Animal and Human Physiology, Faculty of Biology, National and Kapodistrian University of Athens, 15784, Athens, Greece
| | | | - Fabio Di Lisa
- Department of Biomedical Sciences, Università Degli Studi di Padova, Padova, Italy
| | - Efstathios K Iliodromitis
- 2nd Department of Cardiology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece.
| |
Collapse
|
18
|
Mitochondria and Pharmacologic Cardiac Conditioning-At the Heart of Ischemic Injury. Int J Mol Sci 2021; 22:ijms22063224. [PMID: 33810024 PMCID: PMC8004818 DOI: 10.3390/ijms22063224] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Pharmacologic cardiac conditioning increases the intrinsic resistance against ischemia and reperfusion (I/R) injury. The cardiac conditioning response is mediated via complex signaling networks. These networks have been an intriguing research field for decades, largely advancing our knowledge on cardiac signaling beyond the conditioning response. The centerpieces of this system are the mitochondria, a dynamic organelle, almost acting as a cell within the cell. Mitochondria comprise a plethora of functions at the crossroads of cell death or survival. These include the maintenance of aerobic ATP production and redox signaling, closely entwined with mitochondrial calcium handling and mitochondrial permeability transition. Moreover, mitochondria host pathways of programmed cell death impact the inflammatory response and contain their own mechanisms of fusion and fission (division). These act as quality control mechanisms in cellular ageing, release of pro-apoptotic factors and mitophagy. Furthermore, recently identified mechanisms of mitochondrial regeneration can increase the capacity for oxidative phosphorylation, decrease oxidative stress and might help to beneficially impact myocardial remodeling, as well as invigorate the heart against subsequent ischemic insults. The current review highlights different pathways and unresolved questions surrounding mitochondria in myocardial I/R injury and pharmacological cardiac conditioning.
Collapse
|
19
|
Yang X, Zhou Y, Liang H, Meng Y, Liu H, Zhou Y, Huang C, An B, Mao H, Liao Z. VDAC1 promotes cardiomyocyte autophagy in anoxia/reoxygenation injury via the PINK1/Parkin pathway. Cell Biol Int 2021; 45:1448-1458. [PMID: 33675282 DOI: 10.1002/cbin.11583] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/05/2021] [Accepted: 02/27/2021] [Indexed: 12/15/2022]
Abstract
Ischemia/reperfusion (I/R) is a well-known injury to the myocardium, but the mechanism involved remains elusive. In addition to the well-accepted apoptosis theory, autophagy was recently found to be involved in the process, exerting a dual role as protection in ischemia and detriment in reperfusion. Activation of autophagy is mediated by mitochondrial permeability transition pore (MPTP) opening during reperfusion. In our previous study, we showed that MPTP opening is regulated by VDAC1, a channel protein located in the outer membrane of mitochondria. Thus, upregulation of VDAC1 expression is a possible trigger to cardiomyocyte autophagy via an unclear pathway. Here, we established an anoxia/reoxygenation (A/R) model in vitro to simulate the I/R process in vivo. At the end of A/R treatment, VDAC1, Beclin 1, and LC3-II/I were upregulated, and autophagic vacuoles were increased in cardiomyocytes, which showed a connection of VDAC1 and autophagy development. These variations also led to ROS burst, mitochondrial dysfunction, and aggravated apoptosis. Knockdown of VDAC1 by RNAi could alleviate the above-mentioned cellular damages. Additionally, the expression of PINK1 and Parkin was enhanced after A/R injury. Furthermore, Parkin was recruited to mitochondria from the cytosol, which suggested that the PINK1/Parkin autophagic pathway was activated during A/R. Nevertheless, the PINK1/Parkin pathway was effectively inhibited when VDAC1 was knocked-down. Taken together, the A/R-induced cardiomyocyte injury was mediated by VDAC1 upregulation, which led to cell autophagy via the PINK1/Parkin pathway, and finally aggravated apoptosis.
Collapse
Affiliation(s)
- Xiaomei Yang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Yuancheng Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Haiyan Liang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Yan Meng
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Haocheng Liu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Ying Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Chunhong Huang
- Department of Biochemistry, College of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Binyi An
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongli Mao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Zhangping Liao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Ramalingam A, Mohd Fauzi N, Budin SB, Zainalabidin S. Impact of prolonged nicotine administration on myocardial function and susceptibility to ischaemia-reperfusion injury in rats. Basic Clin Pharmacol Toxicol 2021; 128:322-333. [PMID: 32991780 DOI: 10.1111/bcpt.13500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/24/2020] [Accepted: 09/17/2020] [Indexed: 01/01/2023]
Abstract
This study investigated the impact of prolonged nicotine administration on myocardial susceptibility to ischaemia-reperfusion (I/R) injury in a rat model and determined whether nicotine affects mitochondrial reactive oxygen species (ROS) production and permeability transition in rat hearts. Sprague-Dawley rats were administered 0.6 or 1.2 mg/kg nicotine for 28 days, and their hearts were isolated at end-point for assessment of myocardial susceptibility to I/R injury ex vivo. Rat heart mitochondria were also isolated from a subset of rats for analysis of mitochondrial ROS production and permeability transition. Compared to the vehicle controls, rat hearts isolated from nicotine-administered rats exhibited poorer left ventricular function that worsened over the course of I/R. Coronary flow rate was also severely impaired in the nicotine groups at baseline and this worsened after I/R. Nicotine administration significantly increased mitochondrial ROS production and permeability transition relative to the vehicle controls. Interestingly, pre-incubation of isolated mitochondria with ROS scavengers (superoxide dismutase and mitoTEMPO) significantly abolished nicotine-induced increase in mitochondria permeability transition in isolated rat heart mitochondria. Overall, our data showed that prolonged nicotine administration enhances myocardial susceptibility to I/R injury in rats and this is associated with mitochondrial ROS-driven increase in mitochondrial permeability transition.
Collapse
Affiliation(s)
- Anand Ramalingam
- Programme of Biomedical Science, Centre of Toxicology and Health Risk Studies (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Norsyahida Mohd Fauzi
- Centre for Drug and Herbal Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siti Balkis Budin
- Programme of Biomedical Science, Centre of Diagnostic, Therapeutic and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- Programme of Biomedical Science, Centre of Toxicology and Health Risk Studies (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
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: 2.3] [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
|
22
|
Huo J, Lu S, Kwong JQ, Bround MJ, Grimes KM, Sargent MA, Brown ME, Davis ME, Bers DM, Molkentin JD. MCUb Induction Protects the Heart From Postischemic Remodeling. Circ Res 2020; 127:379-390. [PMID: 32299299 PMCID: PMC7367751 DOI: 10.1161/circresaha.119.316369] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
RATIONALE Mitochondrial Ca2+ loading augments oxidative metabolism to match functional demands during times of increased work or injury. However, mitochondrial Ca2+ overload also directly causes mitochondrial rupture and cardiomyocyte death during ischemia-reperfusion injury by inducing mitochondrial permeability transition pore opening. The MCU (mitochondrial Ca2+ uniporter) mediates mitochondrial Ca2+ influx, and its activity is modulated by partner proteins in its molecular complex, including the MCUb subunit. OBJECTIVE Here, we sought to examine the function of the MCUb subunit of the MCU-complex in regulating mitochondria Ca2+ influx dynamics, acute cardiac injury, and long-term adaptation after ischemic injury. METHODS AND RESULTS Cardiomyocyte-specific MCUb overexpressing transgenic mice and Mcub gene-deleted (Mcub-/-) mice were generated to dissect the molecular function of this protein in the heart. We observed that MCUb protein is undetectable in the adult mouse heart at baseline, but mRNA and protein are induced after ischemia-reperfusion injury. MCUb overexpressing mice demonstrated inhibited mitochondrial Ca2+ uptake in cardiomyocytes and partial protection from ischemia-reperfusion injury by reducing mitochondrial permeability transition pore opening. Antithetically, deletion of the Mcub gene exacerbated pathological cardiac remodeling and infarct expansion after ischemic injury in association with greater mitochondrial Ca2+ uptake. Furthermore, hindlimb remote ischemic preconditioning induced MCUb expression in the heart, which was associated with decreased mitochondrial Ca2+ uptake, collectively suggesting that induction of MCUb protein in the heart is protective. Similarly, mouse embryonic fibroblasts from Mcub-/- mice were more sensitive to Ca2+ overload. CONCLUSIONS Our studies suggest that Mcub is a protective cardiac inducible gene that reduces mitochondrial Ca2+ influx and permeability transition pore opening after ischemic injury to reduce ongoing pathological remodeling.
Collapse
Affiliation(s)
- Jiuzhou Huo
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.H., M.J.B., K.M.G., M.A.S., J.D.M.)
| | - Shan Lu
- Department of Pharmacology, University of California, Davis (S.L., D.M.B.)
| | - Jennifer Q Kwong
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (J.Q.K.)
| | - Michael J Bround
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.H., M.J.B., K.M.G., M.A.S., J.D.M.)
| | - Kelly M Grimes
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.H., M.J.B., K.M.G., M.A.S., J.D.M.)
| | - Michelle A Sargent
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.H., M.J.B., K.M.G., M.A.S., J.D.M.)
| | - Milton E Brown
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, GA (M.E.B., M.E.D.)
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, GA (M.E.B., M.E.D.)
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis (S.L., D.M.B.)
| | - Jeffery D Molkentin
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.H., M.J.B., K.M.G., M.A.S., J.D.M.)
- Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, OH (J.D.M.)
| |
Collapse
|
23
|
Boengler K, Schlüter KD, Schermuly RT, Schulz R. Cardioprotection in right heart failure. Br J Pharmacol 2020; 177:5413-5431. [PMID: 31995639 PMCID: PMC7680005 DOI: 10.1111/bph.14992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/04/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023] Open
Abstract
Ischaemic and pharmacological conditioning of the left ventricle is mediated by the activation of signalling cascades, which finally converge at the mitochondria and reduce ischaemia/reperfusion (I/R) injury. Whereas the molecular mechanisms of conditioning in the left ventricle are well characterized, cardioprotection of the right ventricle is principally feasible but less established. Similar to what is known for the left ventricle, a dysregulation in signalling pathways seems to play a role in I/R injury of the healthy and failing right ventricle and in the ability/inability of the right ventricle to respond to a conditioning stimulus. The maintenance of mitochondrial function seems to be crucial in both ventricles to reduce I/R injury. As far as currently known, similar molecular mechanisms mediate ischaemic and pharmacological preconditioning in the left and right ventricles. However, the two ventricles seem to respond differently towards exercise‐induced preconditioning. LINKED ARTICLES This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | | | | | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
24
|
Non-secretory renin reduces oxidative stress and increases cardiomyoblast survival during glucose and oxygen deprivation. Sci Rep 2020; 10:2329. [PMID: 32047214 PMCID: PMC7012910 DOI: 10.1038/s41598-020-59216-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Although the renin-angiotensin system usually promotes oxidative stress and cell death, renin transcripts have been discovered, whose transcription product may be cardioprotective. These transcripts encode a non-secretory renin isoform that is localized in the cytosol and within mitochondria. Here we tested the hypotheses that cytosolic renin [ren(2-9)] expression promotes cell survival under hypoxia and glucose depletion by preserving the mitochondrial membrane potential (∆Ψm) and mitigating the accumulation of ROS. To simulate ischemic insults, we exposed H9c2 cells to glucose deprivation, anoxia or to combined oxygen-glucose deprivation (OGD) for 24 hours and determined renin expression. Furthermore, H9c2 cells transfected with the empty pIRES vector (pIRES cells) or ren(2-9) cDNA-containing vector [ren(2-9) cells] were analyzed for cell death, ∆Ψm, ATP levels, accumulation of ROS, and cytosolic Ca2+ content. In pIRES cells, expression of ren(1A-9) was stimulated under all three ischemia-related conditions. After OGD, the cells lost their ∆Ψm and exhibited enhanced ROS accumulation, increased cytosolic Ca2+ levels, decreased ATP levels as well as increased cell death. In contrast, ren(2-9) cells were markedly protected from these effects. Ren(2-9) appears to represent a protective response to OGD by reducing ROS generation and preserving mitochondrial functions. Therefore, it is a promising new target for the prevention of ischemia-induced myocardial damage.
Collapse
|
25
|
Ren JY, Lin DM, Wang CB, Yang YL, Wang ZQ, Cui BQ, Ma J. Postconditioning Protection Against Myocardiocyte Anoxia/Reoxygenation Injury From Penehyclidine Hydrochloride. Drug Des Devel Ther 2019; 13:3977-3988. [PMID: 32063699 PMCID: PMC6884978 DOI: 10.2147/dddt.s224282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/19/2019] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND/AIMS To investigate the postconditioning protective effect of penehyclidine hydrochloride (PHC) against anoxia/reoxygenation (A/R) injury in H9c2 cells along with the involved mechanism and timing effect. METHODS We divided H9c2 cells into 7 groups: control group, A/R group and PHC+A/R groups at 0 min, 5 mins, 10 mins, 20 mins, 30 mins, respectively (treated with 0.1 μm/L PHC at 0 min, 5 mins, 10 mins, 20 mins, 30 mins after the reoxygenation procedure began). Cell apoptosis, oxidative stress, intracellular Ca2+ concentration, mitochondrial membrane potential and mitochondrial permeability transition pore (MPTP) opening were explored. Bcl-2, Bax, Cyt C, caspase-3 and caspase-9 levels were measured. RESULTS A/R significantly increased both cell injury and cell apoptosis. PHC showed postconditioning protective effect by attenuating superoxide production, decreasing Ca2+ overload, restraining MPTP activities, restoring mitochondrial membrane potential, regulating cell apoptosis proteins and modulation of mitochondrial pathway. Earlier administration of PHC offered greater postconditioning protective effect. CONCLUSION H9c2 cells were protected by PHC from A/R injury regardless of timing of PHC administration (0 min, 5 mins, 10 mins, 20 mins, 30 mins). However, earlier administration of PHC resulted in better PHC postconditioning protection.
Collapse
Affiliation(s)
- JY Ren
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People’s Republic of China
| | - DM Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People’s Republic of China
| | - CB Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People’s Republic of China
| | - YL Yang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People’s Republic of China
| | - ZQ Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People’s Republic of China
| | - BQ Cui
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People’s Republic of China
| | - J Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People’s Republic of China
| |
Collapse
|
26
|
Astaxanthin Inhibits Mitochondrial Permeability Transition Pore Opening in Rat Heart Mitochondria. Antioxidants (Basel) 2019; 8:antiox8120576. [PMID: 31766490 PMCID: PMC6943429 DOI: 10.3390/antiox8120576] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022] Open
Abstract
The mitochondrion is the main organelle of oxidative stress in cells. Increased permeability of the inner mitochondrial membrane is a key phenomenon in cell death. Changes in membrane permeability result from the opening of the mitochondrial permeability transition pore (mPTP), a large-conductance channel that forms after the overload of mitochondria with Ca2+ or in response to oxidative stress. The ketocarotenoid astaxanthin (AST) is a potent antioxidant that is capable of maintaining the integrity of mitochondria by preventing oxidative stress. In the present work, the effect of AST on the functioning of mPTP was studied. It was found that AST was able to inhibit the opening of mPTP, slowing down the swelling of mitochondria by both direct addition to mitochondria and administration. AST treatment changed the level of mPTP regulatory proteins in isolated rat heart mitochondria. Consequently, AST can protect mitochondria from changes in the induced permeability of the inner membrane. AST inhibited serine/threonine protein kinase B (Akt)/cAMP-responsive element-binding protein (CREB) signaling pathways in mitochondria, which led to the prevention of mPTP opening. Since AST improves the resistance of rat heart mitochondria to Ca2+-dependent stress, it can be assumed that after further studies, this antioxidant will be considered an effective tool for improving the functioning of the heart muscle in general under normal and medical conditions.
Collapse
|
27
|
Prokudina E, Naryzhnaya N, Mukhomedzyanov A, Gorbunov A, Zhang Y, Jaggi A, Tsibulnikov S, Nesterov E, Lishmanov Y, Suleiman M, Oeltgen P, Maslov L. Effect of Chronic Continuous Normobaric Hypoxia on Functional State of Cardiac Mitochondria and Tolerance of Isolated Rat Heart to Ischemia and Reperfusion: Role of µ and δ2 Opioid Receptors. Physiol Res 2019; 68:909-920. [DOI: 10.33549/physiolres.933945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic continuous normobaric hypoxia (CNH) increases cardiac tolerance to ischemia/reperfusion injury in vivo and this effect is mediated via µ and δ2 opioid receptors (ORs) activation. CNH has also been shown to be cardioprotective in isolated rat heart. In this study, we hypothesize that this cardioprotective effect of CNH is mediated by activation of µ and δ2 ORs and preservation of mitochondrial function. Hearts from rats adapted to CNH (12 % oxygen) for 3 weeks were extracted, perfused in the Langendorff mode and subjected to 45 min of global ischemia and 30 min of reperfusion. Intervention groups were pretreated for 10 min with antagonists for different OR types: naloxone (300 nmol/l), the selective δ OR antagonist TIPP(ψ) (30 nmol/l), the selective δ1 OR antagonist BNTX (1 nmol/l), the selective δ2 OR antagonist naltriben (1 nmol/l), the selective peptide μ OR antagonist CTAP (100 nmol/l) and the selective κ OR antagonist nor-binaltorphimine (3 nmol/l). Creatine kinase activity in coronary effluent and cardiac contractile function were monitored to assess cardiac injury and functional impairment. Additionally, cardiac tissue was collected to measure ATP and to isolate mitochondria to measure respiration rate and calcium retention capacity. Adaptation to CNH decreased myocardial creatine kinase release during reperfusion and improved the postischemic recovery of contractile function. Additionally, CNH improved mitochondrial state 3 and uncoupled respiration rates, ADP/O, mitochondrial transmembrane potential and calcium retention capacity and myocardial ATP level during reperfusion compared to the normoxic group. These protective effects were completely abolished by naloxone, TIPP(ψ), naltriben, CTAP but not BNTX or nor-binaltorphimine. These results suggest that cardioprotection associated with adaptation to CNH is mediated by µ and δ2 opioid receptors activation and preservation of mitochondrial function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - L.N. Maslov
- Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia.
| |
Collapse
|
28
|
Two pharmacological epoxyeicosatrienoic acid-enhancing therapies are effectively antihypertensive and reduce the severity of ischemic arrhythmias in rats with angiotensin II-dependent hypertension. J Hypertens 2019; 36:1326-1341. [PMID: 29570510 DOI: 10.1097/hjh.0000000000001708] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE We examined the effects of treatment with soluble epoxide hydrolase inhibitor (sEHi) and epoxyeicosatrienoic acids (EETs) analogue (EET-A), given alone or combined, on blood pressure (BP) and ischemia/reperfusion myocardial injury in rats with angiotensin II (ANG II)-dependent hypertension. METHODS Ren-2 transgenic rats (TGR) were used as a model of ANG II-dependent hypertension and Hannover Sprague-Dawley rats served as controls. Rats were treated for 14 days with sEHi or EET-A and BP was measured by radiotelemetry. Albuminuria, cardiac hypertrophy and concentrations of ANG II and EETs were determined. Separate groups were subjected to acute myocardial ischemia/reperfusion injury and the infarct size and ventricular arrhythmias were determined. RESULTS Treatment of TGR with sEHi and EET-A, given alone or combined, decreased BP to a similar degree, reduced albuminuria and cardiac hypertrophy to similar extent; only treatment regimens including sEHi increased myocardial and renal tissue concentrations of EETs. sEHi and EET-A, given alone or combined, suppressed kidney ANG II levels in TGR. Remarkably, infarct size did not significantly differ between TGR and Hannover Sprague-Dawley rats, but the incidence of ischemia-induced ventricular fibrillations was higher in TGR. Application of sEHi and EET-A given alone and combined sEHi and EET-A treatment were all equally effective in reducing life-threatening ventricular fibrillation in TGR. CONCLUSION The findings indicate that chronic treatment with either sEHi or EET-A exerts distinct antihypertensive and antiarrhythmic actions in our ANG II-dependent model of hypertension whereas combined administration of sEHi and EET-A does not provide additive antihypertensive or cardioprotective effects.
Collapse
|
29
|
Naryzhnaya NV, Maslov LN, Oeltgen PR. Pharmacology of mitochondrial permeability transition pore inhibitors. Drug Dev Res 2019; 80:1013-1030. [DOI: 10.1002/ddr.21593] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Natalia V. Naryzhnaya
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Leonid N. Maslov
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Peter R. Oeltgen
- Department of PathologyUniversity of Kentucky College of Medicine Lexington Kentucky
| |
Collapse
|
30
|
Román-Anguiano NG, Correa F, Cano-Martínez A, de la Peña-Díaz A, Zazueta C. Cardioprotective effects of Prolame and SNAP are related with nitric oxide production and with diminution of caspases and calpain-1 activities in reperfused rat hearts. PeerJ 2019; 7:e7348. [PMID: 31392096 PMCID: PMC6673759 DOI: 10.7717/peerj.7348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/25/2019] [Indexed: 01/26/2023] Open
Abstract
Cardiac tissue undergoes changes during ischemia-reperfusion (I-R) that compromise its normal function. Cell death is one of the consequences of such damage, as well as diminution in nitric oxide (NO) content. This signaling molecule regulates the function of the cardiovascular system through dependent and independent effects of cyclic guanosine monophosphate (cGMP). The independent cGMP pathway involves post-translational modification of proteins by S-nitrosylation. Studies in vitro have shown that NO inhibits the activity of caspases and calpains through S-nitrosylation of a cysteine located in their catalytic site, so we propose to elucidate if the regulatory mechanisms of NO are related with changes in S-nitrosylation of cell death proteins in the ischemic-reperfused myocardium. We used two compounds that increase the levels of NO by different mechanisms: Prolame, an amino-estrogenic compound with antiplatelet and anticoagulant effects that induces the increase of NO levels in vivo by activating the endothelial nitric oxide synthase (eNOS) and that has not been tested as a potential inhibitor of apoptosis. On the other hand, S-Nitroso-N-acetylpenicillamine (SNAP), a synthetic NO donor that has been shown to decrease cell death after inducing hypoxia-reoxygenation in cell cultures. Main experimental groups were Control, I-R, I-R+Prolame and I-R+SNAP. Additional groups were used to evaluate the NO action pathways. Contractile function represented as heart rate and ventricular pressure was evaluated in a Langendorff system. Infarct size was measured with 2,3,5-triphenyltetrazolium chloride stain. NO content was determined indirectly by measuring nitrite levels with the Griess reaction and cGMP content was measured by Enzyme-Linked ImmunoSorbent Assay. DNA integrity was evaluated by DNA laddering visualized on an agarose gel and by Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling assay. Activities of caspase-3, caspase-8, caspase-9 and calpain-1 were evaluated spectrophotometrically and the content of caspase-3 and calpain-1 by western blot. S-nitrosylation of caspase-3 and calpain-1 was evaluated by labeling S-nitrosylated cysteines. Our results show that both Prolame and SNAP increased NO content and improved functional recovery in post-ischemic hearts. cGMP-dependent and S-nitrosylation pathways were activated in both groups, but the cGMP-independent pathway was preferentially activated by SNAP, which induced higher levels of NO than Prolame. Although SNAP effectively diminished the activity of all the proteases, a correlative link between the activity of these proteases and S-nitrosylation was not fully established.
Collapse
Affiliation(s)
| | - Francisco Correa
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| | - Aurora de la Peña-Díaz
- Departamento de Biología Molecular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México.,Departamento de Farmacología, Universidad Nacional Autónoma de México, México, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| |
Collapse
|
31
|
Influence of Cardiovascular Risk Factors, Comorbidities, Medication Use and Procedural Variables on Remote Ischemic Conditioning Efficacy in Patients with ST-Segment Elevation Myocardial Infarction. Int J Mol Sci 2019; 20:ijms20133246. [PMID: 31269650 PMCID: PMC6650921 DOI: 10.3390/ijms20133246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/21/2019] [Accepted: 06/28/2019] [Indexed: 12/20/2022] Open
Abstract
Remote ischemic conditioning (RIC) confers cardioprotection in patients with ST-segment elevation myocardial infarction (STEMI). Despite intense research, the translation of RIC into clinical practice remains a challenge. This may, at least partly, be due to confounding factors that may modify the efficacy of RIC. The present review focuses on cardiovascular risk factors, comorbidities, medication use and procedural variables which may modify the efficacy of RIC in patients with STEMI. Findings of such efficacy modifiers are based on subgroup and post-hoc analyses and thus hold risk of type I and II errors. Although findings from studies evaluating influencing factors are often ambiguous, some but not all studies suggest that smoking, non-statin use, infarct location, area-at-risk of infarction, pre-procedural Thrombolysis in Myocardial Infarction (TIMI) flow, ischemia duration and coronary collateral blood flow to the infarct-related artery may influence on the cardioprotective efficacy of RIC. Results from the on-going CONDI2/ERIC-PPCI trial will determine any clinical implications of RIC in the treatment of patients with STEMI and predefined subgroup analyses will give further insight into influencing factors on the efficacy of RIC.
Collapse
|
32
|
Hong XY, Hong X, Gu WW, Lin J, Yin WT. Cardioprotection and improvement in endothelial-dependent vasodilation during late-phase of whole body hypoxic preconditioning in spontaneously hypertensive rats via VEGF and endothelin-1. Eur J Pharmacol 2018; 842:79-88. [PMID: 30401629 DOI: 10.1016/j.ejphar.2018.10.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/17/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022]
Abstract
The present study was designed to investigate the effect of late phase of whole body hypoxic preconditioning on endothelial-dependent vasorelaxation and cardioprotection from ischemia-reperfusion injury in spontaneously hypertensive rats (SHR). Hypoxic preconditioning was performed by subjecting rats to four episodes of alternate exposure to low O2 (8%) and normal air O2 of 10 min each. After 24 h, the mesenteric arteries and hearts were isolated to determine the vascular function and cardioprotection from ischemia-reperfusion (I/R) injury on the Langendorff apparatus. There was a significant impairment in acetylcholine-induced relaxation in norepinephrine precontracted arteries (endothelium-dependent function) and increase in I/R-induced myocardial injury in SHR in comparison to Wistar Kyoto rats (WKY). However, hypoxic preconditioning significantly restored endothelium-dependent relaxation in SHR and attenuated I/R injury in both SHR and WKY. Hypoxic preconditioning also led to an increase in the levels of endothelin-1 (not endothelin-2 or -3), vascular endothelial growth factor-A (VEGF-A) and HIF-1α levels. Pretreatment with bevacizumab (anti-VEGF-A) and bosentan (endothelin receptor blocker) significantly attenuated hypoxic preconditioning-induced restoration of endothelium-dependent relaxation and cardioprotection from I/R injury. These interventions also attenuated the levels of VEGF-A and HIF-1α without modulating the endothelin-1 levels. It may be concluded that an increase in the endothelin-1 levels with a subsequent increase in HIF-1α and VEGF expression may possibly contribute in improving endothelium-dependent vasorelaxation and protecting hearts from I/R injury in SHR during late phase of whole body hypoxic preconditioning.
Collapse
Affiliation(s)
- Xing-Yu Hong
- Department of Vascular Surgery, China-Japan Union Hospital of JiLin University, ChangChun 130031, China.
| | - Xin Hong
- Department of Vascular Surgery, China-Japan Union Hospital of JiLin University, ChangChun 130031, China.
| | - Wei-Wei Gu
- Department of Hepatopancreatobility Surgery, China-Japan Union Hospital of JiLin University, ChangChun 130031, China.
| | - Jie Lin
- Department of Vascular Surgery, China-Japan Union Hospital of JiLin University, ChangChun 130031, China.
| | - Wei-Tian Yin
- Department of Hand Surgery, China-Japan Union Hospital of JiLin University, ChangChun 130031, China.
| |
Collapse
|
33
|
Szobi A, Farkašová‐Ledvényiová V, Lichý M, Muráriková M, Čarnická S, Ravingerová T, Adameová A. Cardioprotection of ischaemic preconditioning is associated with inhibition of translocation of MLKL within the plasma membrane. J Cell Mol Med 2018; 22:4183-4196. [PMID: 29921042 PMCID: PMC6111849 DOI: 10.1111/jcmm.13697] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/26/2018] [Indexed: 12/17/2022] Open
Abstract
Necroptosis, a form of cell loss involving the RIP1-RIP3-MLKL axis, has been identified in cardiac pathologies while its inhibition is cardioprotective. We investigated whether the improvement of heart function because of ischaemic preconditioning is associated with mitigation of necroptotic signaling, and these effects were compared with a pharmacological antinecroptotic approach targeting RIP1. Langendorff-perfused rat hearts were subjected to ischaemic preconditioning with or without a RIP1 inhibitor (Nec-1s). Necroptotic signaling and the assessment of oxidative damage and a putative involvement of CaMKII in this process were analysed in whole tissue and subcellular fractions. Ischaemic preconditioning, Nec-1s and their combination improved postischaemic heart function recovery and reduced infarct size to a similar degree what was in line with the prevention of MLKL oligomerization and translocation to the membrane. On the other hand, membrane peroxidation and apoptosis were unchanged by either approach. Ischaemic preconditioning failed to ameliorate ischaemia-reperfusion-induced increase in RIP1 and RIP3 while pSer229-RIP3 levels were reduced only by Nec-1s. In spite of the additive phosphorylation of CaMKII and PLN because of ditherapy, the postischaemic contractile force and relaxation was comparably improved in all the intervention groups while antiarrhythmic effects were observed in the ischaemic preconditioning group only. Necroptosis inhibition seems to be involved in cardioprotection of ischaemic preconditioning and is comparable but not intensified by an anti-RIP1 agent. Changes in oxidative stress nor CaMKII signaling are unlikely to explain the beneficial effects.
Collapse
Affiliation(s)
- Adrián Szobi
- Faculty of PharmacyComenius University in BratislavaBratislavaSlovakia
| | | | - Martin Lichý
- Faculty of PharmacyComenius University in BratislavaBratislavaSlovakia
| | - Martina Muráriková
- Centre of Experimental MedicineInstitute for Heart ResearchSlovak Academy of SciencesBratislavaSlovakia
| | - Slávka Čarnická
- Centre of Experimental MedicineInstitute for Heart ResearchSlovak Academy of SciencesBratislavaSlovakia
| | - Tatiana Ravingerová
- Centre of Experimental MedicineInstitute for Heart ResearchSlovak Academy of SciencesBratislavaSlovakia
| | - Adriana Adameová
- Faculty of PharmacyComenius University in BratislavaBratislavaSlovakia
| |
Collapse
|
34
|
|
35
|
Hernández-Reséndiz S, Muñoz-Vega M, Contreras WE, Crespo-Avilan GE, Rodriguez-Montesinos J, Arias-Carrión O, Pérez-Méndez O, Boisvert WA, Preissner KT, Cabrera-Fuentes HA. Responses of Endothelial Cells Towards Ischemic Conditioning Following Acute Myocardial Infarction. CONDITIONING MEDICINE 2018; 1:247-258. [PMID: 30338315 PMCID: PMC6191189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
One of the primary therapeutic goals of modern cardiology is to design strategies aimed at minimizing myocardial infarct size and optimizing cardiac function following acute myocardial infarction (AMI). Patients with AMI who underwent reperfusion therapy display dysfunction of the coronary endothelium. Consequently, ischemic endothelial cells become more permeable and weaken their natural anti-thrombotic and anti-inflammatory potential. Ischemia-reperfusion injury (IRI) is associated with activation of the humoral and cellular components of the hemostatic and innate immune system, and also with excessive production of reactive oxygen species (ROS), the inhibition of nitric oxide synthase, and with inflammatory processes. Given its essential role in the regulation of vascular homeostasis, involving platelets and leukocytes among others, dysfunctional endothelium can lead to increased risk of coronary vasospasm and thrombosis. Endothelial dysfunction can be prevented by ischemic conditioning with a protective intervention based on limited intermittent periods of ischemia and reperfusion. The molecular mechanisms and signal transduction pathways underlying conditioning phenomena in the coronary endothelium have been described as involving less ROS production, reduced adhesion of neutrophils to endothelial cells and diminished inflammatory reactions. This review summarizes our current understanding of the cellular and molecular mechanisms regulating IRI-affected and -damaged coronary endothelium, and how ischemic conditioning may preserve its function.
Collapse
Affiliation(s)
- Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
| | - Mónica Muñoz-Vega
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González. Ciudad de México, México
| | - Whendy E Contreras
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | | | - Oscar Arias-Carrión
- Molecular Biology Department, Instituto Nacional de Cardiología "Ignacio Chávez", C.D de México, México
| | - Oscar Pérez-Méndez
- Molecular Biology Department, Instituto Nacional de Cardiología "Ignacio Chávez", C.D de México, México
| | - William A Boisvert
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Hawaii, USA
| | - Klaus T Preissner
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
36
|
Rosenberg JH, Werner JH, Moulton MJ, Agrawal DK. Current Modalities and Mechanisms Underlying Cardioprotection by Ischemic Conditioning. J Cardiovasc Transl Res 2018; 11:292-307. [PMID: 29797232 PMCID: PMC6117210 DOI: 10.1007/s12265-018-9813-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023]
Abstract
Ischemic preconditioning is a process which serves to mitigate reperfusion injury. Preconditioning of the heart can be achieved through natural, pharmacological, and mechanical means. Mechanical preconditioning appears to have the greatest chance of good outcomes while methods employing pharmacologic preconditioning have been largely unsuccessful. Remote ischemic preconditioning achieves a cardioprotective effect by applying cycles of ischemia and reperfusion in a distal limb, stimulating the release of a neurohumoral cardioprotective factor incited by stimulation of afferent neurons. The cardioprotective factor stimulates the reperfusion injury salvage kinase (RISK) and survivor activator factor enhancement (SAFE) signaling cascades in cardiomyocytes which promote cell survival by the expression of anti-apoptotic genes and inhibition of the opening of mitochondrial permeability transition pores. Clinical application of ischemic preconditioning involving targets in the RISK and SAFE signaling appears promising in the treatment of acute myocardial infarction; however, clinical trials have yet to demonstrate additional benefit to current therapy.
Collapse
Affiliation(s)
- John H Rosenberg
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - John H Werner
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Michael J Moulton
- Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
37
|
Parviz Y, Waleed M, Vijayan S, Adlam D, Lavi S, Al Nooryani A, Iqbal J, Stone GW. Cellular and molecular approaches to enhance myocardial recovery after myocardial infarction. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2018; 20:351-364. [PMID: 29958820 DOI: 10.1016/j.carrev.2018.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/22/2018] [Accepted: 05/29/2018] [Indexed: 10/14/2022]
Abstract
Reperfusion therapy has resulted in significant improvement in post-myocardial infarction morbidity and mortality in over the last 4 decades. Nonetheless, it is well recognized that simply restoring patency of the epicardial artery may not stop or reverse damage at microvascular level, and myocardial salvage is often suboptimal. Numerous efforts have been undertaken to elucidate the mechanisms underlying extensive myonecrosis to facilitate the discovery of therapies to provide additional and incremental benefits over current therapeutic pathways. To date, conclusively effective strategies to promote myocardial recovery have not yet been established. Novel approaches are investigating the foundational cellular and molecular bases of myocardial ischemia and irreversible injury. Herein, we review the emerging concepts and proposed therapies that may improve myocardial protection and reduce infarct size. We examine the preclinical and clinical evidence for reduced infarct size with these strategies, including anti-inflammatory agents, intracellular ion channel modulators, agents affecting the reperfusion injury salvage kinase (RISK) and nitric oxide signaling pathways, modulators of mitochondrial function, anti-apoptotic agents, and stem cell and gene therapy. We review the potential reasons of failures to date and the potential for new strategies to further promote myocardial recovery and improve prognosis.
Collapse
Affiliation(s)
- Yasir Parviz
- New York Presbyterian Hospital, Columbia University Medical Centre and the Cardiovascular Research Foundation, New York, NY, USA.
| | | | | | - David Adlam
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, UK
| | - Shahar Lavi
- Division of Cardiology, London Health Sciences Centre, Western University, London, Ontario, Canada
| | | | - Javaid Iqbal
- South Yorkshire Cardiothoracic Centre, Northern General Hospital, Sheffield, UK
| | - Gregg W Stone
- New York Presbyterian Hospital, Columbia University Medical Centre and the Cardiovascular Research Foundation, New York, NY, USA
| |
Collapse
|
38
|
Zhao W, Li S, Ren C, Meng R, Ji X. Chronic Remote Ischemic Conditioning May Mimic Regular Exercise:Perspective from Clinical Studies. Aging Dis 2018; 9:165-171. [PMID: 29392091 PMCID: PMC5772854 DOI: 10.14336/ad.2017.1015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/15/2017] [Indexed: 11/01/2022] Open
Abstract
Chronic remote ischemic conditioning (RIC), particularly long-term repeated RIC, has been applied in clinical trials with the expectation that it could play its protective roles for protracted periods. In sports medicine, chronic RIC has also been demonstrated to improve exercise performance, akin to improvements seen with regular exercise training. Therefore, chronic RIC may mimic regular exercise, and they may have similar underlying mechanisms. In this study, we explored the common underlying mechanisms of chronic RIC and physical exercise in protecting multiple organs and benefiting various populations, the advantages of chronic RIC, and the challenges for its popularization. Intriguingly, several underlying mechanisms of RIC and exercise have been shown to overlap. These include the production of many autacoids, enhanced ability for antioxidant activity, modulating immune and inflammatory responses. Therefore, it appears that chronic RIC, just like regular exercise, has beneficial effects in unhealthy, sub-healthy and healthy individuals. Compared with regular exercise, chronic RIC has several advantages, which may provide novel insights into the area of exercise and health. Chronic RIC may enrich the modes of exercise, and benefit individuals with severe diseases. Also, the disabled, and sub-healthy individuals are likely to benefit from chronic RIC either as an alternative to exercise or an adjunct to pharmacological or non-pharmacological therapy.
Collapse
Affiliation(s)
- Wenbo Zhao
- ¹Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- 2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,3Beijing Municipal Geriatric Medical Research Center, Beijing, China.,4National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Changhong Ren
- 2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,3Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Ran Meng
- ¹Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- 2Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,4National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
39
|
Myofibrillogenesis Regulator 1 Rescues Renal Ischemia/Reperfusion Injury by Recruitment of PI3K-Dependent P-AKT to Mitochondria. Shock 2018; 46:531-540. [PMID: 27219857 DOI: 10.1097/shk.0000000000000658] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To investigate whether myofibrillogenesis regulator 1 (MR-1) attenuates renal ischemia/reperfusion (I/R) injury via inhibiting phosphorylated Akt (p-Akt) mitochondrial translocation-mediated opening of the mitochondrial permeability transition pore (mPTP), we injected adenovirus containing MR-1 gene or its siRNAs to the left kidney subcapsular areas of Sprague-Dawley rats, which subsequently underwent experimental renal I/R injury. Renal functions and the severity of the tubular injury were evaluated by the serum creatinine and blood urea nitrogen levels and the pathological scores. We also examined the mitochondrial morphology and functions. Total/p-Akt were assessed by western blot using the mitochondrial and the cytosolic fractions of cortex of renal tissue, respectively. We found that mitochondrial and cytosolic MR-1 levels and mitochondrial p-Akt decreased, and cytosolic p-Akt increased after reperfusion. Subcapsular injection of adenovirus led to higher MR-1 expression in the mitochondria/cytosol, inhibited mPTP opening, and alleviated renal I/R injury; adenovirus injection also upregulated mitochondrial total and p-Akt levels more prominently compared with the normal saline (NS) group. Subcapsular injection of MR-1 siRNAs significantly lowered MR-1 expression and induced renal injury, with increased mPTP opening and mitochondrial damage, similar to I/R injury. MR-1 interacted with Akt in renal cortex homogenate. Wortmannin, a phosphatidylinositol 3 kinase (PI3K) inhibitor, abolished both mitochondrial p-Akt recruitment and the protective effect of MR-1 overexpression on I/R injury. To conclude, MR-1 protects kidney against I/R injury through inhibiting mPTP opening and maintaining mitochondrial integrity, through the recruitment of PI3K-dependent p-Akt to the mitochondria. MR-1 could be a new therapeutic strategy for renal I/R injury.
Collapse
|
40
|
Kawai S, Yamada T, Matsuura T, Funao T, Nishikawa K. Neuropathic pain attenuates ischemia reperfusion injury through β2-adrenergic pathway. Life Sci 2017; 187:9-16. [PMID: 28827152 DOI: 10.1016/j.lfs.2017.08.002] [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: 05/20/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 12/23/2022]
Abstract
AIMS The relationship between neuropathic pain and myocardial infarction (MI) was uncertain because of some medication or underlying diseases. This study investigated the impact of neuropathic pain on ischemia reperfusion injury using isolated rat hearts and cardiomyocytes. MAIN METHODS Male Sprague-Dawley rats were assigned to the control and allodynia (AL) groups, with the latter subjected to the fifth lumbar spinal-nerve ligation. First, isolated hearts underwent 25-min ischemia and 90-min reperfusion to assess hemodynamic changes and MI area. Second, isolated cardiomyocytes underwent 10-min laser illumination to assess the opening of mitochondrial permeability transition pore (mPTP) and cellular hypercontraction. Lastly, expression of pro-survival kinases was measured in another cardiomyocytes using flow cytometry. AL-treated hearts were concomitantly examined regarding the involvement of β-adrenergic pathways by esmolol (ESM), β1-blocker (100μM, AL+ESM), and ICI118551 (ICI), β2-blocker (50nM, AL+ICI). KEY FINDINGS All hemodynamic variables did not change significantly in between-group comparisons except at 30min of reperfusion. MI area decreased remarkably in the AL and AL+ESM groups after 90-min reperfusion. The AL+ICI group significantly increased it as compared with the AL and AL+ESM groups. Similarly, the AL and AL+ESM groups significantly inhibited mPTP opening and cellular hypercontraction, whereas the AL+ICI group reversed these effects. Enhanced expression of pro-survival kinases was observed in the AL and AL+ESM groups, but the AL+ICI group abolished this enhancement. SIGNIFICANCE Our findings suggested that neuropathic pain possessed cardioprotective effects through inhibiting mPTP opening. The underlying mechanisms were possibly regulated by β2-adrenergic activation and pro-survival kinase expression in cardiomyocytes.
Collapse
Affiliation(s)
- Shigeaki Kawai
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| | - Tokuhiro Yamada
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan.
| | - Tadashi Matsuura
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| | - Tomoharu Funao
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| | - Kiyonobu Nishikawa
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| |
Collapse
|
41
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
42
|
|
43
|
Mitochondria as a target of cardioprotection in models of preconditioning. J Bioenerg Biomembr 2017; 49:357-368. [PMID: 28730272 DOI: 10.1007/s10863-017-9720-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 06/14/2017] [Indexed: 12/24/2022]
Abstract
Over the recent years the view on mitochondria in the heart as a cellular powerhouse providing ATP supply needed to sustain contractile function, basal metabolic processes, and ionic homeostasis has changed radically. At present it is known that dysfunctions of these organelles are essential in the development of a large number of diseases, including cardiovascular diseases. Moreover, mitochondria are considered to be a very promising target of endogenous strategies that are essential in the protection of the myocardium from acute ischemia/reperfusion injury. These strategies including ischemic preconditioning, remote ischemic preconditioning as well as the acute phase of streptozotocin-induced diabetes mellitus, provide a similar effect of protection. Alterations observed in the functional and structural properties of heart mitochondria caused by short-term pathological impulses are associated with endogenous cardioprotective processes. It seems that the extent of mitochondrial membrane fluidization could be an active response mechanism to injury with a subtle effect on membrane-associated processes which further affect the environment of the whole organelle, thus inducing metabolic changes in the heart. In this review article, we provide an overview of endogenous protective mechanisms induced by hypoxic, pseudohypoxic and ischemic conditions with special consideration of the role of heart mitochondria in these processes.
Collapse
|
44
|
Harisseh R, Chiari P, Villedieu C, Sueur P, Abrial M, Fellahi JL, Ovize M, Gharib A. Cyclophilin D Modulates the Cardiac Mitochondrial Target of Isoflurane, Sevoflurane, and Desflurane. J Cardiovasc Pharmacol 2017; 69:326-334. [PMID: 28328748 DOI: 10.1097/fjc.0000000000000479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Volatile anesthetics are known to limit myocardial ischemia-reperfusion injuries. Mitochondria were shown to be major contributors to cardioprotection. Cyclophilin D (CypD) is one of the main regulators of mitochondria-induced cell death. We compared the effect of isoflurane, sevoflurane, and desflurane in the presence or absence of CypD, to clarify its role in the mechanism of cardioprotection induced by these anesthetics. METHODS Oxidative phosphorylation, mitochondrial membrane potential, and H2O2 production were measured in isolated mitochondria from wild-type (WT) or CypD knockout mice in basal conditions and after hypoxia-reoxygenation in the presence or absence of volatile anesthetics. RESULTS All volatile anesthetics inhibited mitochondrial state 3 of complex I, decreased membrane potential, and increased adenosine diphosphate consumption duration in both WT and CypD knockout mice. However, they differently modified H2O2 production after stimulation by succinate: CypD ablation reduced H2O2 production, isoflurane decreased H2O2 level in WT but not in CypD knockout mice, sevoflurane affected both lines whereas desflurane increased H2O2 production in CypD knockout and had no effect on WT mice. CONCLUSIONS This study showed different effects of isoflurane, sevoflurane, and desflurane on mitochondrial functions and highlighted the implication of CypD in the regulation of adenosine diphosphate consumption and complex I-induced radical oxygen species production.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Anesthetics, Inhalation/pharmacology
- Animals
- Peptidyl-Prolyl Isomerase F
- Cyclophilins/deficiency
- Cyclophilins/genetics
- Cyclophilins/metabolism
- Cytoprotection
- Desflurane
- Energy Metabolism/drug effects
- Genotype
- Hydrogen Peroxide/metabolism
- Isoflurane/analogs & derivatives
- Isoflurane/pharmacology
- Male
- Membrane Potential, Mitochondrial/drug effects
- Methyl Ethers/pharmacology
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Myocardial Reperfusion Injury/enzymology
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxidative Phosphorylation/drug effects
- Phenotype
- Protective Agents/pharmacology
- Sevoflurane
- Time Factors
Collapse
Affiliation(s)
- Rania Harisseh
- *INSERM UMR 1060, CarMeN Laboratory, Univ Lyon1, IHU OPERA, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France; †Service d'Anesthésie Réanimation, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France; and ‡Service d'Explorations Fonctionnelles Cardiovasculaires & CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Lizano P, Rashed E, Stoll S, Zhou N, Wen H, Hays TT, Qin G, Xie LH, Depre C, Qiu H. The valosin-containing protein is a novel mediator of mitochondrial respiration and cell survival in the heart in vivo. Sci Rep 2017; 7:46324. [PMID: 28425440 PMCID: PMC5397870 DOI: 10.1038/srep46324] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/16/2017] [Indexed: 12/24/2022] Open
Abstract
The valosin-containing protein (VCP) participates in signaling pathways essential for cell homeostasis in multiple tissues, however, its function in the heart in vivo remains unknown. Here we offer the first description of the expression, function and mechanism of action of VCP in the mammalian heart in vivo in both normal and stress conditions. By using a transgenic (TG) mouse with cardiac-specific overexpression (3.5-fold) of VCP, we demonstrate that VCP is a new and powerful mediator of cardiac protection against cell death in vivo, as evidenced by a 50% reduction of infarct size after ischemia/reperfusion versus wild type. We also identify a novel role of VCP in preserving mitochondrial respiration and in preventing the opening of mitochondrial permeability transition pore in cardiac myocytes under stress. In particular, by genetic deletion of inducible isoform of nitric oxide synthase (iNOS) from VCP TG mouse and by pharmacological inhibition of iNOS in isolated cardiac myocytes, we reveal that an increase of expression and activity of iNOS in cardiomyocytes by VCP is an essential mechanistic link of VCP-mediated preservation of mitochondrial function. These data together demonstrate that VCP may represent a novel therapeutic avenue for the prevention of myocardial ischemia.
Collapse
Affiliation(s)
- Paulo Lizano
- Department of Cell Biology and Molecular Medicine; New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Eman Rashed
- Department of Cell Biology and Molecular Medicine; New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Shaunrick Stoll
- Division of Physiology, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA, 92324, USA
| | - Ning Zhou
- Division of Physiology, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA, 92324, USA
| | - Hairuo Wen
- Department of Cell Biology and Molecular Medicine; New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Tristan T Hays
- Division of Physiology, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA, 92324, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB),Birmingham, AL, 35294, USA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine; New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Christophe Depre
- Department of Cell Biology and Molecular Medicine; New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Hongyu Qiu
- Department of Cell Biology and Molecular Medicine; New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.,Division of Physiology, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA, 92324, USA
| |
Collapse
|
46
|
Guerrero-Orriach JL, Escalona Belmonte JJ, Ramirez Fernandez A, Ramirez Aliaga M, Rubio Navarro M, Cruz Mañas J. Cardioprotection with halogenated gases: how does it occur? Drug Des Devel Ther 2017; 11:837-849. [PMID: 28352158 PMCID: PMC5358986 DOI: 10.2147/dddt.s127916] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Numerous studies have studied the effect of halogenated agents on the myocardium, highlighting the beneficial cardiac effect of the pharmacological mechanism (preconditioning and postconditioning) when employed before and after ischemia in patients with ischemic heart disease. Anesthetic preconditioning is related to the dose-dependent signal, while the degree of protection is related to the concentration of the administered drug and the duration of the administration itself. Triggers for postconditioning and preconditioning might have numerous pathways in common; mitochondrial protection and a decrease in inflammatory mediators could be the major biochemical elements. Several pathways have been identified, including attenuation of NFκB activation and reduced expression of TNFα, IL-1, intracellular adhesion molecules, eNOS, the hypercontraction reduction that follows reperfusion, and antiapoptotic activating kinases (Akt, ERK1/2). It appears that the preconditioning and postconditioning triggers have numerous similar paths. The key biochemical elements are protection of the mitochondria and reduction in inflammatory mediators, both of which are developed in various ways. We have studied this issue, and have published several articles on cardioprotection with halogenated gases. Our results confirm greater cardioprotection through myocardial preconditioning in patients anesthetized with sevoflurane compared with propofol, with decreasing levels of troponin and N-terminal brain natriuretic peptide prohormone. The difference between our studies and previous studies lies in the use of sedation with sevoflurane in the postoperative period. The results could be related to a prolonged effect, in addition to preconditioning and postconditioning, which could enhance the cardioprotective effect of sevoflurane in the postoperative period. With this review, we aim to clarify the importance of various mechanisms involved in preconditioning and postconditioning with halogenated gases, as supported by our studies.
Collapse
Affiliation(s)
- Jose Luis Guerrero-Orriach
- Department of Cardioanesthesiology, Virgen de la Victoria University Hospital
- Instituto de Investigación Biomédica de Málaga (IBIMA)
- Department of Pharmacology and Pediatrics, University of Malaga, Malaga, Spain
| | | | | | | | | | - Jose Cruz Mañas
- Department of Cardioanesthesiology, Virgen de la Victoria University Hospital
| |
Collapse
|
47
|
Abe JI, Sandhu UG, Hoang NM, Thangam M, Quintana-Quezada RA, Fujiwara K, Le NT. Coordination of Cellular Localization-Dependent Effects of Sumoylation in Regulating Cardiovascular and Neurological Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:337-358. [PMID: 28197922 PMCID: PMC5716632 DOI: 10.1007/978-3-319-50044-7_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sumoylation, a reversible post-transcriptional modification process, of proteins are involved in cellular differentiation, growth, and even motility by regulating various protein functions. Sumoylation is not limited to cytosolic proteins as recent evidence shows that nuclear proteins, those associated with membranes, and mitochondrial proteins are also sumoylated. Moreover, it is now known that sumoylation plays an important role in the process of major human ailments such as malignant, cardiovascular and neurological diseases. In this chapter, we will highlight and discuss how the localization of SUMO protease and SUMO E3 ligase in different compartments within a cell regulates biological processes that depend on sumoylation. First, we will discuss the key role of sumoylation in the nucleus, which leads to the development of endothelial dysfunction and atherosclerosis . We will then discuss how sumoylation of plasma membrane potassium channel proteins are involved in epilepsy and arrhythmia. Mitochondrial proteins are known to be also sumoylated, and the importance of dynamic-related protein 1 (DRP1) sumoylation on mitochondrial function will be discussed. As we will emphasize throughout this review, sumoylation plays crucial roles in different cellular compartments, which is coordinately regulated by the translocation of various SUMO proteases and SUMO E3 ligase. Comprehensive approach will be necessary to understand the molecular mechanism for efficiently moving around various enzymes that regulate sumoylation within cells.
Collapse
Affiliation(s)
- Jun-Ichi Abe
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA.
| | - Uday G Sandhu
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Nguyet Minh Hoang
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Manoj Thangam
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Raymundo A Quintana-Quezada
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Keigi Fujiwara
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Nhat Tu Le
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| |
Collapse
|
48
|
Connexin 43 and Mitochondria in Cardiovascular Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:227-246. [PMID: 28551790 DOI: 10.1007/978-3-319-55330-6_12] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Connexin 43 (Cx43) is the major connexin protein in ventricular cardiomyocytes. Six Cx43 proteins assemble into so-called hemichannels at the sarcolemma and opposing hemichannels form gap junctions, which allow the passage of small molecules and electrical current flow between adjacent cells. Apart from its localization at the plasma membrane, Cx43 is also present in cardiomyocyte mitochondria, where it is important for mitochondrial function in terms of oxygen consumption and potassium fluxes. The expression of gap junctional and mitochondrial Cx43 is altered under several pathophysiological conditions among them are hypertension, hypertrophy, hypercholesterolemia, ischemia/reperfusion injury, post-infarction remodeling, and heart failure. The present review will focus on the role of Cx43 in cardiovascular diseases and will highlight the importance of mitochondrial Cx43 in cardioprotection.
Collapse
|
49
|
Andrienko T, Pasdois P, Rossbach A, Halestrap AP. Real-Time Fluorescence Measurements of ROS and [Ca2+] in Ischemic / Reperfused Rat Hearts: Detectable Increases Occur only after Mitochondrial Pore Opening and Are Attenuated by Ischemic Preconditioning. PLoS One 2016; 11:e0167300. [PMID: 27907091 PMCID: PMC5131916 DOI: 10.1371/journal.pone.0167300] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/12/2016] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial permeability transition pore (mPTP) opening is critical for ischemia / reperfusion (I/R) injury and is associated with increased [Ca2+] and reactive oxygen species (ROS). Here we employ surface fluorescence to establish the temporal sequence of these events in beating perfused hearts subject to global I/R. A bespoke fluorimeter was used to synchronously monitor surface fluorescence and reflectance of Langendorff-perfused rat hearts at multiple wavelengths, with simultaneous measurements of hemodynamic function. Potential interference by motion artefacts and internal filtering was assessed and minimised. Re-oxidation of NAD(P)H and flavoproteins on reperfusion (detected using autofluorescence) was rapid (t0.5 < 15 s) and significantly slower following ischemic preconditioning (IP). This argues against superoxide production from reduced Complex 1 being a critical mediator of initial mPTP opening during early reperfusion. Furthermore, MitoPY1 (a mitochondria-targeted H2O2-sensitive fluorescent probe) and aconitase activity measurements failed to detect matrix ROS increases during early reperfusion. However, two different fluorescent cytosolic ROS probes did detect ROS increases after 2–3 min of reperfusion, which was shown to be after initiation of mPTP opening. Cyclosporin A (CsA) and IP attenuated these responses and reduced infarct size. [Ca2+]i (monitored with Indo-1) increased progressively during ischemia, but dropped rapidly within 90 s of reperfusion when total mitochondrial [Ca2+] was shown to be increased. These early changes in [Ca2+] were not attenuated by IP, but substantial [Ca2+] increases were observed after 2–3 min reperfusion and these were prevented by both IP and CsA. Our data suggest that the major increases in ROS and [Ca2+] detected later in reperfusion are secondary to mPTP opening. If earlier IP-sensitive changes occur that might trigger initial mPTP opening they are below our limit of detection. Rather, we suggest that IP may inhibit initial mPTP opening by alternative mechanisms such as prevention of hexokinase 2 dissociation from mitochondria during ischemia.
Collapse
Affiliation(s)
- Tatyana Andrienko
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Philippe Pasdois
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
- INSERM U1045—L'Institut de Rythmologie et Modélisation Cardiaque (LIRYC), Université de Bordeaux, Bordeaux, France
| | - Andreas Rossbach
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Andrew P Halestrap
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Hausenloy DJ, Barrabes JA, Bøtker HE, Davidson SM, Di Lisa F, Downey J, Engstrom T, Ferdinandy P, Carbrera-Fuentes HA, Heusch G, Ibanez B, Iliodromitis EK, Inserte J, Jennings R, Kalia N, Kharbanda R, Lecour S, Marber M, Miura T, Ovize M, Perez-Pinzon MA, Piper HM, Przyklenk K, Schmidt MR, Redington A, Ruiz-Meana M, Vilahur G, Vinten-Johansen J, Yellon DM, Garcia-Dorado D. Ischaemic conditioning and targeting reperfusion injury: a 30 year voyage of discovery. Basic Res Cardiol 2016; 111:70. [PMID: 27766474 PMCID: PMC5073120 DOI: 10.1007/s00395-016-0588-8] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 10/11/2016] [Indexed: 01/12/2023]
Abstract
To commemorate the auspicious occasion of the 30th anniversary of IPC, leading pioneers in the field of cardioprotection gathered in Barcelona in May 2016 to review and discuss the history of IPC, its evolution to IPost and RIC, myocardial reperfusion injury as a therapeutic target, and future targets and strategies for cardioprotection. This article provides an overview of the major topics discussed at this special meeting and underscores the huge importance and impact, the discovery of IPC has made in the field of cardiovascular research.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK. .,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK. .,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore. .,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.
| | - Jose A Barrabes
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, 8200, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Fabio Di Lisa
- Department of Biomedical Sciences and CNR Institute of Neurosciences, University of Padova, Padua, Italy
| | - James Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Thomas Engstrom
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Hector A Carbrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Institute for Biochemistry, Medical Faculty Justus-Liebig-University, Giessen, Germany.,Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Gerd Heusch
- Institute for Pathophysiology, West-German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Efstathios K Iliodromitis
- 2nd University Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Javier Inserte
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain
| | | | - Neena Kalia
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Rajesh Kharbanda
- Oxford Heart Centre, The John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Faculty of Health Sciences, University of Cape Town, Chris Barnard Building, Anzio Road, Observatory, Cape Town, Western Cape, 7925, South Africa
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St. Thomas' Hospital, London, UK
| | - Tetsuji Miura
- Department of Cardiovascular, Renal, and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Lyon, France.,UMR 1060 (CarMeN), Université Claude Bernard, Lyon 1, France
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Hans Michael Piper
- Carl von Ossietzky Universität Oldenburg, Ökologiezentrum, Raum 2-116, Uhlhornsweg 99 b, 26129, Oldenburg, Germany
| | - Karin Przyklenk
- Department of Physiology and Emergency Medicine, Cardiovascular Research Institute, Wayne State University, Detroit, MI, USA
| | - Michael Rahbek Schmidt
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
| | - Andrew Redington
- Division of Cardiology, Department of Pediatrics, Heart Institute, Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Marisol Ruiz-Meana
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Research Center, CSIC-ICCC, IIB-Hospital Sant Pau, c/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
| | - Jakob Vinten-Johansen
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University, Atlanta, USA
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - David Garcia-Dorado
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain.
| |
Collapse
|