1
|
Doul J, Minaříková M, Charvátová Z, Maxová H. Nitric oxide is involved in the cardioprotection of neonatal rat hearts, but not in neonatal ischemic postconditioning. Physiol Rep 2024; 12:e16147. [PMID: 39097984 PMCID: PMC11298247 DOI: 10.14814/phy2.16147] [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: 02/17/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
The cardioprotective effect of ischemic preconditioning (IPC) and ischemic postconditioning (IPoC) in adult hearts is mediated by nitric oxide (NO). During the early developmental period, rat hearts exhibit higher resistance to ischemia-reperfusion (I/R) injury, contain higher levels of serum nitrates, and their resistance cannot be further increased by IPC or IPoC. NOS blocker (L-NAME) lowers their high resistance. Wistar rat hearts (postnatal Days 1 and 10) were perfused according to Langendorff and exposed to 40 min of global ischemia followed by reperfusion with or without IPoC. NO and reactive oxygen species donors (DEA-NONO, SIN-1) and L-NAME were administered. Tolerance to ischemia decreased between Days 1 and 10. DEA-NONO (low concentrations) significantly increased tolerance to I/R injury on both Days 1 and 10. SIN-1 increased tolerance to I/R injury on Day 10, but not on Day 1. L-NAME significantly reduced resistance to I/R injury on Day 1, but actually increased resistance to I/R injury on Day 10. Cardioprotection by IPoC on Day 10 was not affected by either NO donors or L-NAME. It can be concluded that resistance of the neonatal heart to I/R injury is NO dependent, but unlike in adult hearts, cardioprotective interventions, such as IPoC, are most likely NO independent.
Collapse
Affiliation(s)
- Jan Doul
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Marcela Minaříková
- Department of Physiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Zuzana Charvátová
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Hana Maxová
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
- Center for Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| |
Collapse
|
2
|
Jiang L, Zhou X, Zhao X, Wang Z, Huang A, Huang Y, Sun H, Guan F, Jiang W. Tetrandrine downregulates TRPV2 expression to ameliorate myocardial ischemia/reperfusion injury in rats via regulation of cardiomyocyte apoptosis, calcium homeostasis and mitochondrial function. Eur J Pharmacol 2024; 964:176246. [PMID: 38061472 DOI: 10.1016/j.ejphar.2023.176246] [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: 05/30/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
Our previous study has indicated that tetrandrine (TET) can target miR-202-5p to repress the activation of transient receptor potential vanilloid type 2 (TRPV2), eventually ameliorating the progression of myocardial ischemia/reperfusion injury (MI/RI). This study is aimed to further ascertain the detailed mechanisms between TET and TRPV2 in MI/RI pathogenesis. Here, a myocardial I/R injury rat model and a hypoxia-reoxygenation (H/R) model in rat myocardial cell line (H9C2 cells) were established. We reported that pronounced upregulation of TRPV2 was observed in I/R rats and H/R-induced H9C2 cells. Silencing of TRPV2 could improve cardiac function and myocardial injury, reduced infarction size, and promoted cardiomyocyte proliferation in I/R rats. In I/R rats or H/R-induced H9C2 cells, cardiomyocyte apoptosis was inhibited by knocking-down TRPV2. Meanwhile, the silenced TRPV2 or TET treatment ameliorated the damaged mitochondrial structure, mitigated ROS generation, restored the impaired ΔΨM, inhibited mPTP opening and alleviated Ca2+ overload in H/R-induced H9C2 cells. The results obtained from the overexpression of TRPV2 were contrary to those depicted above. Moreover, TET could downregulate TRPV2 expression, while the overexpression of TRPV2 could reverse the above protective effects of TET in H/R-induced H9C2 cells. The results indicated that TET may function as a TRPV2 blocking agent, thereby attenuating the progression of MI/RI through modulation of cardiomyocyte apoptosis, calcium homeostasis and mitochondrial function. These findings offer a theoretical foundation for potential clinical application of TET therapy in patients with MI/RI.
Collapse
Affiliation(s)
- Lelin Jiang
- The Second Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Xue Zhou
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Xiaoli Zhao
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Zhaolin Wang
- The Medical College of Shanghai University, Shanghai, 200000, China.
| | - Anwu Huang
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Yiwei Huang
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Huanghui Sun
- Department of Heart Function Examination, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Fanlu Guan
- Department of Cardiology, The Third Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Wenbing Jiang
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
3
|
Yang Y, Qi J, Zhang M, Chen P, Liu Y, Sun X, Chu L. The cardioprotective effects and mechanisms of naringenin in myocardial ischemia based on network pharmacology and experiment verification. Front Pharmacol 2022; 13:954555. [PMID: 36160433 PMCID: PMC9500410 DOI: 10.3389/fphar.2022.954555] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Naringenin (Nar) is a natural flavonoid extracted from citrus fruits with abundant pharmacological properties against cardiac diseases, but existing studies are unsystematic and scattered. The present research systematically investigates the mechanism of action of Nar in the treatment of myocardial ischemia (MI). Network pharmacology was used to analyze the relevant targets of Nar against MI as well as the biological mechanisms. The protective effect of Nar was initially assessed in H9c2 cells induced by CoCl2. In acutely isolated rat cardiomyocytes, Nar was further explored for effects on L-type Ca2+ currents, cell contractility and Ca2+ transients by using patch-clamp technique and Ion Optix system. Network pharmacology analysis indicated that Nar improved apoptosis, mitochondrial energy metabolism, inflammation and oxidative stress. Experimental validation demonstrated that Nar decreased ROS and MDA levels and increased antioxidant activity (e.g., GSH-PX, SOD, and CAT), mitochondrial membrane potential, ATP and Ca2+-ATPase contents. Nar also markedly reduced inflammatory factor levels, apoptosis, and intracellular Ca2+ concentrations in H9c2 cells. Based on the experimental results, it is speculated that Ca2+ signals play an essential role in the process of Nar against MI. Thus, we further confirmed that Nar significantly inhibited the L-type Ca2+ currents, contractility and Ca2+ transients in acutely isolated cardiomyocytes. The inhibition of Ca2+ overload by Nar may be a novel cardioprotective mechanism. The present study may serve as a basis for future clinical research, and Nar as a Ca2+ channel inhibitor may provide new perspectives for the treatment of myocardial ischemic diseases.
Collapse
Affiliation(s)
- Yakun Yang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jiaying Qi
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Muqing Zhang
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Pingping Chen
- College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yanshuang Liu
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- *Correspondence: Yanshuang Liu, ; Xiaorun Sun, ; Li Chu,
| | - Xiaorun Sun
- College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- *Correspondence: Yanshuang Liu, ; Xiaorun Sun, ; Li Chu,
| | - Li Chu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- *Correspondence: Yanshuang Liu, ; Xiaorun Sun, ; Li Chu,
| |
Collapse
|
4
|
Coutinho DCO, Santos-Miranda A, Joviano-Santos JV, Foureaux G, Santos A, Rodrigues-Ferreira C, Martins-Júnior PA, Resende RR, Medei E, Vieyra A, Santos RAS, Cruz JS, Ferreira AJ. Diminazene Aceturate, an angiotensin converting enzyme 2 (ACE2) activator, promotes cardioprotection in ischemia/reperfusion-induced cardiac injury. Peptides 2022; 151:170746. [PMID: 35033621 DOI: 10.1016/j.peptides.2022.170746] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 01/03/2023]
Abstract
This study aimed to investigate whether the Diminazene Aceturate (DIZE), an angiotensin-converting enzyme 2 (ACE2) activator, can revert cardiac dysfunction in ischemia reperfusion-induced (I/R) injury in animals and examine the mechanism underlying this effect. Wistar rats systemically received DIZE (1 mg/kg) for thirty days. Cardiac function in isolated rat hearts was evaluated using the Langendorff technique. After I/R, ventricular non-I/R and I/R samples were used to evaluate ATP levels. Mitochondrial function was assessed using cardiac permeabilized fibers and isolated cardiac mitochondria. Cardiac cellular electrophysiology was evaluated using the patch clamp technique. DIZE protected the heart after I/R from arrhythmia and cardiac dysfunction by preserving ATP levels, independently of any change in coronary flow and heart rate. DIZE improved mitochondrial function, increasing the capacity for generating ATP and reducing proton leak without changing the specific citrate synthase activity. The activation of the ACE2 remodeled cardiac electrical profiles, shortening the cardiac action potential duration at 90 % repolarization. Additionally, cardiomyocytes from DIZE-treated animals exhibited reduced sensibility to diazoxide (KATP agonist) and a higher KATP current compared to the controls. DIZE was able to improve mitochondrial function and modulate cardiac electrical variables with a cardio-protective profile, resulting in direct myocardial cell protection from I/R injury.
Collapse
Affiliation(s)
| | - Artur Santos-Miranda
- Laboratory of CardioBiology, Department of Biophysics, Federal University of Sao Paulo, Brazil
| | | | - Giselle Foureaux
- Department of Morphology, Federal University of Minas Gerais, Brazil
| | - Anderson Santos
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Brazil
| | - Clara Rodrigues-Ferreira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo A Martins-Júnior
- Department of Child and Adolescent Oral Health, Federal University of Minas Gerais, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Brazil
| | - Emiliano Medei
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson A S Santos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics, Federal University of Minas Gerais, Brazil
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Brazil
| | | |
Collapse
|
5
|
Maslov LN, Popov SV, Mukhomedzyanov AV, Naryzhnaya NV, Voronkov NS, Ryabov VV, Boshchenko AA, Khaliulin I, Prasad NR, Fu F, Pei JM, Logvinov SV, Oeltgen PR. Reperfusion Cardiac Injury: Receptors and the Signaling Mechanisms. Curr Cardiol Rev 2022; 18:63-79. [PMID: 35422224 PMCID: PMC9896422 DOI: 10.2174/1573403x18666220413121730] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/01/2022] [Accepted: 01/10/2022] [Indexed: 11/22/2022] Open
Abstract
It has been documented that Ca2+ overload and increased production of reactive oxygen species play a significant role in reperfusion injury (RI) of cardiomyocytes. Ischemia/reperfusion induces cell death as a result of necrosis, necroptosis, apoptosis, and possibly autophagy, pyroptosis and ferroptosis. It has also been demonstrated that the NLRP3 inflammasome is involved in RI of the heart. An increase in adrenergic system activity during the restoration of coronary perfusion negatively affected cardiac resistance to RI. Toll-like receptors are involved in RI of the heart. Angiotensin II and endothelin-1 aggravated ischemic/reperfusion injury of the heart. Activation of neutrophils, monocytes, CD4+ T-cells and platelets contributes to cardiac ischemia/reperfusion injury. Our review outlines the role of these factors in reperfusion cardiac injury.
Collapse
Affiliation(s)
- Leonid N. Maslov
- Address correspondence to this author at the Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Kyevskskaya 111A, 634012 Tomsk, Russia; Tel. +7 3822 262174; E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Menezes-Rodrigues FS, Tavares JGP, Vasques ER, Errante PR, Araújo EAD, Pires-Oliveira M, Scorza CA, Scorza FA, Taha MO, Caricati-Neto A. Cardioprotective effects of pharmacological blockade of the mitochondrial calcium uniporter on myocardial ischemia-reperfusion injury. Acta Cir Bras 2020; 35:e202000306. [PMID: 32692797 PMCID: PMC7251977 DOI: 10.1590/s0102-865020200030000006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/22/2020] [Indexed: 05/13/2023] Open
Abstract
PURPOSE To evaluate whether the attenuation of mitochondrial Ca2+ overload produced by pharmacological blockade of mitochondrial Ca2+ uniporter (MCU) protects the myocardium against injuries caused by cardiac ischemia and reperfusion (CIR). METHODS CIR was induced in adult male Wistar rats (300-350 g) by occlusion of the left anterior descendent coronary artery (10 min), followed by reperfusion (120 min). Rats were treated with different doses of MCU blocker ruthenium red (RuR), administered 5 min before ischemia or reperfusion. RESULTS In untreated rats, the incidences of ventricular arrhythmias (VA), atrioventricular block (AVB) and the lethality (LET) induced by CIR were 85%, 79% and 70%, respectively. In rats treated with RuR before ischemia, the incidences of VA, AVB and LET were significantly reduced to 62%, 25% and 25%, respectively. In rats treated with RuR after ischemia, the incidences of VA, AVB and LET were significantly reduced to 50%, 25% and 25%, respectively. CONCLUSION The significant reduction of the incidence of CIR-induced VA, AVB and LET produced by the treatment with RuR indicates that the attenuation of mitochondrial Ca2+ overload produced by pharmacological blockade of MCU can protect the myocardium against injuries caused by CIR.
Collapse
|
7
|
Wu L, Tan JL, Chen ZY, Huang G. Cardioprotection of post-ischemic moderate ROS against ischemia/reperfusion via STAT3-induced the inhibition of MCU opening. Basic Res Cardiol 2019; 114:39. [DOI: 10.1007/s00395-019-0747-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 08/19/2019] [Indexed: 12/20/2022]
|
8
|
Recent Advances in Pharmacological and Non-Pharmacological Strategies of Cardioprotection. Int J Mol Sci 2019; 20:ijms20164002. [PMID: 31426434 PMCID: PMC6720817 DOI: 10.3390/ijms20164002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemic heart diseases (IHD) are the leading cause of death worldwide. Although the principal form of treatment of IHD is myocardial reperfusion, the recovery of coronary blood flow after ischemia can cause severe and fatal cardiac dysfunctions, mainly due to the abrupt entry of oxygen and ionic deregulation in cardiac cells. The ability of these cells to protect themselves against injury including ischemia and reperfusion (I/R), has been termed “cardioprotection”. This protective response can be stimulated by pharmacological agents (adenosine, catecholamines and others) and non-pharmacological procedures (conditioning, hypoxia and others). Several intracellular signaling pathways mediated by chemical messengers (enzymes, protein kinases, transcription factors and others) and cytoplasmic organelles (mitochondria, sarcoplasmic reticulum, nucleus and sarcolemma) are involved in cardioprotective responses. Therefore, advancement in understanding the cellular and molecular mechanisms involved in the cardioprotective response can lead to the development of new pharmacological and non-pharmacological strategies for cardioprotection, thus contributing to increasing the efficacy of IHD treatment. In this work, we analyze the recent advances in pharmacological and non-pharmacological strategies of cardioprotection.
Collapse
|
9
|
Li Y, Li Q, Zhang O, Guan X, Xue Y, Li S, Zhuang X, Zhou B, Miao G. miR-202-5p protects rat against myocardial ischemia reperfusion injury by downregulating the expression of Trpv2 to attenuate the Ca 2+ overload in cardiomyocytes. J Cell Biochem 2019; 120:13680-13693. [PMID: 31062423 DOI: 10.1002/jcb.28641] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND This study was aimed to unveil micro RNA (miRNA) expression profiles in myocardial ischemia-reperfusion (MI/R) rats and explore whether and how dysregulated miRNAs were involved in the initiation and progression of MI/R in a calcium-dependent manner. METHOD AND RESULTS Rat model of MI/R was established and cardiomyocytes isolated from neonatal rats cardiomyocytes were induced. Both miRNA and messenger RNA expression profiles were analyzed by Microarray. Quantitative reverse-transcription polymerase chain reaction, immunoblotting, bioinformatics analysis, dual-luciferase reporter gene assay, hematoxylin and eosin, Evans blue, and triphenyl tetrazolium chloride were also used in this study. Serum concentrations of myocardial enzymes (phosphocreatine kinase [CK], creatine kinase [CK-MB], lactate dehydrogenase [LDH]), cardiomyocytes loadage of Ca2+ , as well as the expression level of inositol 1,4,5-trisphosphate receptors (IP3R) and sarcoplasmic reticulum Ca2+ -ATPase 2a (SERCA2a) were measured, respectively. Effects of upregulation or downregulation of miR-202-5p or Trpv2 on these indicators were investigated in vivo and in vitro. In MI/R rats and hypoxia/reoxygenation-induced NCMs, miR-202-5p was downregulated, while Trpv2 was upregulated. Trpv2 was a promising target of miR-202-5p and negatively regulated by miR-202-5p. Upregulation of miR-202-5p or downregulation of Trpv2 significantly reduced the serum concentration of myocardial enzymes, as well as cardiomyocyte-produced reactive oxygen species, but inhibition of miR-202-5p or overexpression of Trpv2 brought the worsening situation for these indicators. Besides, upregulation of miR-202-5p upregulation or downregulation of Trpv2 also inhibited Ca2+ overload in cardiomyocytes, accompanied with the increase of SERCA2a and suppression of IP3R. The reduced damage degree and infarct size in myocardial tissue were contrarily worsened by miR-202-5p inhibitor. CONCLUSION Overexpression of miR-202-5p or downregulation of its downstream Trpv2 presented the cardioprotective effects to MI/R rats.
Collapse
Affiliation(s)
- Yanbing Li
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qiang Li
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ou Zhang
- Department of Cardiology, Beijing Tsinghua Chang Gung Hospital, Tsinghua University, Beijing, China
| | - Xiaonan Guan
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yajun Xue
- Department of Cardiology, Beijing Tsinghua Chang Gung Hospital, Tsinghua University, Beijing, China
| | - Siyuan Li
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xianjing Zhuang
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Boda Zhou
- Department of Cardiology, Beijing Tsinghua Chang Gung Hospital, Tsinghua University, Beijing, China
| | - Guobin Miao
- Department of Cardiology, Beijing Tsinghua Chang Gung Hospital, Tsinghua University, Beijing, China
| |
Collapse
|
10
|
Oil recovery performance of a modified HAPAM with lower hydrophobicity, higher molecular weight: A comparative study with conventional HAPAM, HPAM. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2018.12.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
Lesnefsky EJ, Chen Q, Tandler B, Hoppel CL. Mitochondrial Dysfunction and Myocardial Ischemia-Reperfusion: Implications for Novel Therapies. Annu Rev Pharmacol Toxicol 2017; 57:535-565. [PMID: 27860548 PMCID: PMC11060135 DOI: 10.1146/annurev-pharmtox-010715-103335] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondria have emerged as key participants in and regulators of myocardial injury during ischemia and reperfusion. This review examines the sites of damage to cardiac mitochondria during ischemia and focuses on the impact of these defects. The concept that mitochondrial damage during ischemia leads to cardiac injury during reperfusion is addressed. The mechanisms that translate ischemic mitochondrial injury into cellular damage, during both ischemia and early reperfusion, are examined. Next, we discuss strategies that modulate and counteract these mechanisms of mitochondrial-driven injury. The new concept that mitochondria are not merely stochastic sites of oxidative and calcium-mediated injury but that they activate cellular responses of mitochondrial remodeling and cellular reactions that modulate the balance between cell death and recovery is reviewed, and the therapeutic implications of this concept are discussed.
Collapse
Affiliation(s)
- Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia 23298; ,
- Medical Service, McGuire Veterans Affairs Medical Center, Richmond, Virginia 23249;
| | - Qun Chen
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia 23298; ,
| | - Bernard Tandler
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, Ohio 44106;
| | - Charles L Hoppel
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106;
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
- Center for Mitochondrial Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| |
Collapse
|
12
|
Namekata I, Hamaguchi S, Iida-Tanaka N, Kusakabe T, Kato K, Kawanishi T, Tanaka H. Fluorescence Analysis of the Mitochondrial Effect of a Plasmalemmal Na +/Ca 2+ Exchanger Inhibitor, SEA0400, in Permeabilized H9c2 Cardiomyocytes. Biol Pharm Bull 2017; 40:1551-1555. [DOI: 10.1248/bpb.b17-00079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Iyuki Namekata
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Shogo Hamaguchi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Naoko Iida-Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
- Department of Food Science, Otsuma Women’s University
| | - Taichi Kusakabe
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, Toho University
| | - Keisuke Kato
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, Toho University
| | | | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| |
Collapse
|
13
|
de Lima Portella R, Lynn Bickta J, Shiva S. Nitrite Confers Preconditioning and Cytoprotection After Ischemia/Reperfusion Injury Through the Modulation of Mitochondrial Function. Antioxid Redox Signal 2015; 23:307-27. [PMID: 26094636 DOI: 10.1089/ars.2015.6260] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Nitrite is now recognized as an intrinsic signaling molecule that mediates a number of biological processes. One of the most reproducible effects of nitrite is its ability to mediate cytoprotection after ischemia/reperfusion (I/R). This robust phenomenon has been reproduced by a number of investigators in varying animal models focusing on different target organs. Furthermore, nitrite's cytoprotective versatility is highlighted by its ability to mediate delayed preconditioning and remote conditioning in addition to acute protection. RECENT ADVANCES In the last 10 years, significant progress has been made in elucidating the mechanisms underlying nitrite-mediated ischemic tolerance. CRITICAL ISSUES The mitochondrion, which is essential to both the progression of I/R injury and the protection afforded by preconditioning, has emerged as a major subcellular target for nitrite. This review will outline the role of the mitochondrion in I/R injury and preconditioning, review the accumulated preclinical studies demonstrating nitrite-mediated cytoprotection, and finally focus on the known interactions of nitrite with mitochondria and their role in the mechanism of nitrite-mediated ischemic tolerance. FUTURE DIRECTIONS These studies set the stage for current clinical trials testing the efficacy of nitrite to prevent warm and cold I/R injury.
Collapse
Affiliation(s)
- Rafael de Lima Portella
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Janelle Lynn Bickta
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
14
|
Chen Y, Liu J, Zheng Y, Wang J, Wang Z, Gu S, Tan J, Jing Q, Yang H. Uncoupling protein 3 mediates H₂O₂ preconditioning-afforded cardioprotection through the inhibition of MPTP opening. Cardiovasc Res 2014; 105:192-202. [PMID: 25514931 DOI: 10.1093/cvr/cvu256] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIMS Uncoupling protein 3 (UCP3), located in the mitochondrial inner membrane, is cardioprotective, but its mechanisms of preserving mitochondrial function during ischaemia/reperfusion (I/R) are not fully understood. This study investigated whether UCP3 mediates/mimics the cardioprotection of H₂O₂ preconditioning (H₂O₂PC) against I/R injury and the downstream pathway that mediates H₂O₂PC- and UCP3-afforded cardioprotection. METHODS AND RESULTS H₂O₂PC at 20 µM for 5 min significantly improved post-ischaemic functional recovery and reduced lactate dehydrogenase (LDH) release and infarct size with concurrently up-regulated UCP3 expressions in perfused rat hearts subjected to global no-flow I/R. These protections were blocked by UCP3 knockdown with short hairpin RNA but mimicked by UCP3 overexpression. Consistently, H₂O₂PC-attenuated I/R-induced cytosolic and mitochondrial Ca(2+) overload, Ca(2+) transient suppression, mitochondrial reactive oxygen species burst, and loss of mitochondrial inner membrane potential were reversed by UCP3 knockdown but mimicked by UCP3 overexpression. Moreover, co-immunoprecipitation assay revealed an interaction of UCP3 with the mitochondrial permeability transition pore (mPTP) component, adenine nucleotide translocator (ANT), while the cardioprotection induced by H₂O₂PC- and UCP3 overexpression in mitochondria, cardiac function, and cell survival was abolished by atractyloside, a mPTP opener binding to ANT, and partially inhibited by a PI3K/Akt inhibitor wortmannin. Furthermore, H₂O₂PC up-regulated the phosphorylation of Akt, and glycogen synthase kinase 3β was blocked by UCP3 knockdown but mimicked by UCP3 overexpression. CONCLUSION UCP3 mediates the cardioprotection of H₂O₂PC against I/R injury by preserving the mitochondrial function through inhibiting mPTP opening via the interaction with ANT and the PI3K/Akt pathway. Our findings reveal novel mechanisms of UCP3 in the cardioprotection.
Collapse
Affiliation(s)
- Yixiong Chen
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Jinlong Liu
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Yanjun Zheng
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Jinxi Wang
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Zhihua Wang
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Shanshan Gu
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Jiliang Tan
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Qing Jing
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| | - Huangtian Yang
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), 320 Yue Yang Road, Biological Research Building A, Shanghai 200031, China
| |
Collapse
|
15
|
Leu S, Kao YH, Sun CK, Lin YC, Tsai TH, Chang LT, Chua S, Yeh KH, Wu CJ, Fu M, Yip HK. Myocardium-derived conditioned medium improves left ventricular function in rodent acute myocardial infarction. J Transl Med 2011; 9:11. [PMID: 21244680 PMCID: PMC3033820 DOI: 10.1186/1479-5876-9-11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 01/18/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We investigated whether myocardium-derived conditioned medium (MDCM) is effective in preserving left ventricular (LV) function in a rat acute myocardial infarction (AMI) model. METHODS Adult male Sprague-Dawley (SD) rats (n = 36) randomized to receive either left coronary artery ligation (AMI induction) or thoracotomy only (sham procedure) were grouped as follows (n = 6 per group): Group I, II, and III were sham-controls treated by fresh medium, normal rat MDCM, and infarct-related MDCM, respectively. Group IV, V, and VI were AMI rats treated by fresh medium, normal MDCM, and infarct-related MDCM, respectively. Either 75 μL MDCM or fresh medium was administered into infarct myocardium, followed by intravenous injection (3 mL) at postoperative 1, 12, and 24 h. RESULTS In vitro studies showed higher phosphorylated MMP-2 and MMP-9, but lower α-smooth muscle actin and collagen expressions in neonatal cardiac fibroblasts treated with MDCM compared with those in the cardiac fibroblasts treated with fresh medium (all p < 0.05). Sirius-red staining showed larger collagen deposition area in LV myocardium in Group IV than in other groups (all p < 0.05). Stromal cell-derived factor-1α and CXCR4 protein expressions were higher in Group VI than in other groups (all p < 0.05). The number of von Willebrand factor- and BrdU-positive cells and small vessels in LV myocardium as well as 90-day LV ejection fraction were higher, whereas oxidative stress was lower in Group VI than in Group IV and Group V (all p < 0.05). CONCLUSION MDCM therapy reduced cardiac fibrosis and oxidative stress, enhanced angiogenesis, and preserved 90-day LV function in a rat AMI model.
Collapse
Affiliation(s)
- Steve Leu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Shih RH, Cheng SE, Tung WH, Yang CM. Up-Regulation of Heme Oxygenase-1 Protects Against Cold Injury-Induced Brain Damage: A Laboratory-Based Study. J Neurotrauma 2010; 27:1477-87. [DOI: 10.1089/neu.2009.1201] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Ruey-Horng Shih
- Department of Pharmacology, Chang Gung University, Tao-Yuan, Taiwan
| | - Shin-Ei Cheng
- Department of Pharmacology, Chang Gung University, Tao-Yuan, Taiwan
| | - Wei-Hsuan Tung
- Department of Pharmacology, Chang Gung University, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Pharmacology, Chang Gung University, Tao-Yuan, Taiwan
| |
Collapse
|
17
|
Halestrap AP, Pasdois P. The role of the mitochondrial permeability transition pore in heart disease. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1787:1402-15. [PMID: 19168026 DOI: 10.1016/j.bbabio.2008.12.017] [Citation(s) in RCA: 277] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 12/19/2008] [Accepted: 12/20/2008] [Indexed: 01/17/2023]
Abstract
Like Dr. Jeckyll and Mr. Hyde, mitochondria possess two distinct persona. Under normal physiological conditions they synthesise ATP to meet the energy needs of the beating heart. Here calcium acts as a signal to balance the rate of ATP production with ATP demand. However, when the heart is overloaded with calcium, especially when this is accompanied by oxidative stress, mitochondria embrace their darker side, and induce necrotic cell death of the myocytes. This happens acutely in reperfusion injury and chronically in congestive heart failure. Here calcium overload, adenine nucleotide depletion and oxidative stress combine forces to induce the opening of a non-specific pore in the mitochondrial membrane, known as the mitochondrial permeability transition pore (mPTP). The molecular nature of the mPTP remains controversial but current evidence implicates a matrix protein, cyclophilin-D (CyP-D) and two inner membrane proteins, the adenine nucleotide translocase (ANT) and the phosphate carrier (PiC). Inhibition of mPTP opening can be achieved with inhibitors of each component, but targeting CyP-D with cyclosporin A (CsA) and its non-immunosuppressive analogues is the best described. In animal models, inhibition of mPTP opening by either CsA or genetic ablation of CyP-D provides strong protection from both reperfusion injury and congestive heart failure. This confirms the mPTP as a promising drug target in human cardiovascular disease. Indeed, the first clinical trials have shown CsA treatment improves recovery after treatment of a coronary thrombosis with angioplasty.
Collapse
Affiliation(s)
- Andrew P Halestrap
- Department of Biochemistry and Bristol Heart Institute, University of Bristol, School of Medical Sciences, University Walk, Bristol BS8 1TD, UK.
| | | |
Collapse
|
18
|
Ross JL, Howlett SE. Beta-adrenoceptor stimulation exacerbates detrimental effects of ischemia and reperfusion in isolated guinea pig ventricular myocytes. Eur J Pharmacol 2008; 602:364-72. [PMID: 19056376 DOI: 10.1016/j.ejphar.2008.11.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 10/20/2008] [Accepted: 11/17/2008] [Indexed: 10/21/2022]
Abstract
We investigated whether beta-adrenoceptor stimulation exacerbates detrimental effects of ischemia and reperfusion on electrical and contractile function and on intracellular Ca(2+) homeostasis in isolated guinea pig ventricular myocytes. Myocytes were exposed to 20 min of simulated ischemia (37 degrees C) in the absence or presence of isoproterenol (10 nM, applied prior to and during ischemia) and reperfused with Tyrode's solution for 30 min. Unloaded cell shortening, Ca(2+) transients (fura-2), and cell viability were recorded at 5 min intervals in field-stimulated cells (2 Hz). In experiments using microelectrodes, membrane potentials, contractions, and transmembrane currents also were recorded at 5 min intervals. In the absence of ischemia, 10 nM isoproterenol had little effect on either contractile function or Ca(2+) homeostasis. In contrast, when cells were exposed to ischemia, isoproterenol increased the size of contractions and Ca(2+) transients and augmented the increase in diastolic Ca(2+) concentration during ischemia in field-stimulated myocytes. Exposure to isoproterenol also promoted contractile depression in reperfusion. In voltage clamp experiments, isoproterenol abolished the decrease in the magnitude of L-type Ca(2+) current caused by ischemia. Isoproterenol also increased the incidence of abnormal contractile activity and induced delayed afterdepolarizations and the arrhythmogenic transient inward current in ischemia. Additionally, the decline in cell viability in ischemia and reperfusion was exacerbated by isoproterenol. This study shows that beta-adrenoceptor stimulation strongly potentiates adverse effects of ischemia and reperfusion on electrical and contractile function. These adverse effects of isoproterenol are likely caused by an increase in intracellular Ca(2+) accumulation during ischemia.
Collapse
Affiliation(s)
- Jenna L Ross
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | |
Collapse
|
19
|
Chen HT, Yang CX, Li H, Zhang CJ, Wen XJ, Zhou J, Fan YL, Huang T, Zeng YM. Cardioprotection of sevoflurane postconditioning by activating extracellular signal-regulated kinase 1/2 in isolated rat hearts. Acta Pharmacol Sin 2008; 29:931-41. [PMID: 18664326 DOI: 10.1111/j.1745-7254.2008.00824.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM The activation of extracellular signal-regulated kinase (ERK)1/2 protects against ischemic-reperfusion injury. Whether ERK1/2 mediates the cardioprotection of sevoflurane postconditioning is unknown. We tested whether sevoflurane postconditioning produces cardioprotection via an ERK1/2-dependent mechanism. METHODS In protocol 1, Langendorff-perfused Sprague-Dawley rat hearts (n=84, 12 per group), with the exception of the Sham group, were subjected to 30 min ischemia followed by 90 min reperfusion and were assigned to the untreated (control) group, followed by 4 cycles of ischemic postconditioning (25 s of each), 3% (v/v) sevoflurane postconditioning (for 5 min and 10 min of washout), and the PD98059 solvent DMSO (<0.2%), ERK1/2 inhibitor PD98059 (20 micromol/L), and Sevo+PD administration. Left ventricular hemodynamics and coronary flow at 30 min of equilibrium were recorded at 30, 60, and 90 min of reperfusion, respectively. Acute infarct size was measured by triphenyltetrazolium chloride staining. The configuration of mitochondria was observed by an electron microscope. Western blot analysis was used to determine the contents of cytosolic and mitochondrial cytochrome c at the end of reperfusion. In protocol 2, after 15 min of reperfusion, the expression of total and phosphorylated forms of ERK1/2 and its downstream target p70S6K was determined by Western blotting. RESULTS No differences in baseline hemodynamics were observed among the experimental groups (P>0.05). After reperfusion, compared with the control group, sevoflurane postconditioning and ischemic postconditioning significantly(P<0.05) improved functional recovery and largely (P<0.05) decreased myocardial infarct size (22.9%+/-4.6% and 21.2%+/-3.8%, vs 39.4%+/- 5.7%, both P<0.05). Sevoflurane-mediated protection was abolished by PD98059. CONCLUSION Anesthetic postconditioning by sevoflurane effectively protects against reperfusion damage by activating ERK1/2 in vitro.
Collapse
Affiliation(s)
- Hong-tao Chen
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical College, Xuzhou 221002, China
| | | | | | | | | | | | | | | | | |
Collapse
|