1
|
Gong G, Wan W, Zhang X, Chen X, Yin J. Management of ROS and Regulatory Cell Death in Myocardial Ischemia-Reperfusion Injury. Mol Biotechnol 2025; 67:1765-1783. [PMID: 38852121 DOI: 10.1007/s12033-024-01173-y] [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: 12/13/2023] [Accepted: 04/02/2024] [Indexed: 06/10/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is fatal to patients, leading to cardiomyocyte death and myocardial remodeling. Reactive oxygen species (ROS) and oxidative stress play important roles in MIRI. There is a complex crosstalk between ROS and regulatory cell deaths (RCD) in cardiomyocytes, such as apoptosis, pyroptosis, autophagy, and ferroptosis. ROS is a double-edged sword. A reasonable level of ROS maintains the normal physiological activity of myocardial cells. However, during myocardial ischemia-reperfusion, excessive ROS generation accelerates myocardial damage through a variety of biological pathways. ROS regulates cardiomyocyte RCD through various molecular mechanisms. Targeting the removal of excess ROS has been considered an effective way to reverse myocardial damage. Many studies have applied antioxidant drugs or new advanced materials to reduce ROS levels to alleviate MIRI. Although the road from laboratory to clinic has been difficult, many scholars still persevere. This article reviews the molecular mechanisms of ROS inhibition to regulate cardiomyocyte RCD, with a view to providing new insights into prevention and treatment strategies for MIRI.
Collapse
Affiliation(s)
- Ge Gong
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Wenhui Wan
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xinghu Zhang
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xiangxuan Chen
- Department of Cardiology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
| | - Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Jiangsu Medical Vocational College, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Nanjing Medical University Kangda College, Nanjing, 211100, China.
| |
Collapse
|
2
|
Xia J, Huang J, Yan Y, Jian C, He J, Wang N, Shi L, Ding Q, Tian H, Gao W. Bifunctional Nanostarch against Neuronal Apoptosis via Mitochondria Protection for Ameliorating Ischemic Stroke Injury and Promoting Long-Term Neurological Recovery. ACS APPLIED BIO MATERIALS 2025. [PMID: 40289357 DOI: 10.1021/acsabm.4c02005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Mitochondrial damage occurs as an initial event following ischemic onset, and the extent of mitochondrial dysfunction is highly correlated to the severity of ischemia-induced cell death. Once appropriate therapeutic interventions are provided, the ischemic tissue can be salvaged, which is of great significance in achieving better neurological outcomes. Herein, we developed a nanosized starch as a targeting nanoplatform, featuring effective blood-brain barrier (BBB) penetration through lactoferrin-mediated transcytosis. Notably, the nanostarch-based delivery of Mdivi-1 and Alda-1 enables controlled release in the acidic lysosome of neurons, effectively inhibiting the pathological mitochondrial fission and metabolizing toxic aldehydes, thereby creating protective effects on maintaining mitochondrial function. Moreover, we demonstrated that mitochondrial protection induces a transition from activated pro-death responses to a pro-survival state by reducing the release of pro-apoptotic proteins, significantly contributing to the long-term recovery of neurological function. Overall, our nanostarch provided an in-depth understanding of the delivery of mitochondrial protectants and underscored the potential and utility of mitochondrial protection for ischemic stroke via minimizing neuronal apoptosis.
Collapse
Affiliation(s)
- Ji Xia
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Huang
- Department of Anesthesiology, Xi'an Central Hospital, Xi'an 710003, China
| | - Yixiao Yan
- Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi, China
| | - Chenxin Jian
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jiansheng He
- Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi, China
| | - Nisha Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Lei Shi
- Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi, China
| | - Qiyang Ding
- Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi, China
| | - Hao Tian
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wei Gao
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
3
|
Lang A, Oehler D, Benkhoff M, Reinders Y, Barcik M, Shahrjerdi K, Kaldirim M, Sickmann A, Dannenberg L, Polzin A, Pfeiler S, Kelm M, Grandoch M, Jung C, Gerdes N. Mitochondrial Creatine Kinase 2 (Ckmt2) as a Plasma-Based Biomarker for Evaluating Reperfusion Injury in Acute Myocardial Infarction. Biomedicines 2024; 12:2368. [PMID: 39457679 PMCID: PMC11504053 DOI: 10.3390/biomedicines12102368] [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: 09/06/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Acute myocardial infarction (AMI), characterized by irreversible heart muscle damage and impaired cardiac function caused by myocardial ischemia, is a leading cause of global mortality. The damage associated with reperfusion, particularly mitochondrial dysfunction and reactive oxygen species (ROS) formation, has emerged as a crucial factor in the pathogenesis of cardiac diseases, leading to the recognition of mitochondrial proteins as potential markers for myocardial damage. This study aimed to identify differentially expressed proteins based on the type of cardiac injury, in particular those with and without reperfusion. METHODS Male C57Bl/6J mice were either left untreated, sham-operated, received non-reperfused AMI, or reperfused AMI. Twenty-four hours after the procedures, left ventricular (LV) function and morphological changes including infarct size were determined using echocardiography and triphenyl tetrazolium chloride (TTC) staining, respectively. In addition, plasma was isolated and subjected to untargeted mass spectrometry and, further on, the ELISA-based validation of candidate proteins. RESULTS We identified mitochondrial creatine kinase 2 (Ckmt2) as a differentially regulated protein in plasma of mice with reperfused but not non-reperfused AMI. Elevated levels of Ckmt2 were significantly associated with infarct size and impaired LV function following reperfused AMI, suggesting a specific involvement in reperfusion damage. CONCLUSIONS Our study highlights the potential of plasma Ckmt2 as a biomarker for assessing reperfusion injury and its impact on cardiac function and morphology in the acute phase of MI.
Collapse
Affiliation(s)
- Alexander Lang
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Daniel Oehler
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Marcel Benkhoff
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Yvonne Reinders
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., 44139 Dortmund, Germany; (Y.R.); (A.S.)
| | - Maike Barcik
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Khatereh Shahrjerdi
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Madlen Kaldirim
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., 44139 Dortmund, Germany; (Y.R.); (A.S.)
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Lisa Dannenberg
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Amin Polzin
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany;
| | - Susanne Pfeiler
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
| | - Malte Kelm
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany;
| | - Maria Grandoch
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany;
- Institute for Translational Pharmacology, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany;
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (A.L.); (D.O.); (M.B.); (M.B.); (K.S.); (M.K.); (L.D.); (A.P.); (S.P.); (M.K.); (C.J.)
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany;
| |
Collapse
|
4
|
Guo B, Song H, Fan J, Wang B, Chen L, Hu Q, Yin Y. The NR2B-targeted intervention alleviates the neuronal injuries at the sub-acute stage of cerebral ischemia: an exploration of stage-dependent strategy against ischemic insults. Exp Brain Res 2023; 241:2735-2750. [PMID: 37845379 DOI: 10.1007/s00221-023-06717-3] [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: 03/28/2023] [Accepted: 09/30/2023] [Indexed: 10/18/2023]
Abstract
Stroke is reported to be the second leading cause of death worldwide, among which ischemic stroke has fourfold greater incidence than intracerebral hemorrhage. Excitotoxicity induced by NMDAR plays a central role in ischemic stroke-induced neuronal death. However, intervention targeted NMDARs against ischemic stroke has failed, which may result from the complex composition of NMDARs and the dynamic changes of their subunits. In this current study, the levels of NR1, NR2A and NR2B subunits of NMDARs were observed upon different time points during the reperfusion after 1 h ischemia with the western blot assay. It was found that the changes of NR1 subunit were only detected after ischemia 1 h/reperfusion 1 day (1 d). While, the changes of NR2A and NR2B subunits may last to ischemia 1 h/reperfusion 7 day(7 d), indicating that NR2subunits may be a potential target for ischemia-reperfusion injuries at the sub-acute stage of ischemic stroke. Simultaneously, mitochondrial injuries in neurons were investigated with transmission electron microscopy (TEM), and mitochondrial dysfunction was evaluated with mitochondrial membrane proteins oxidative respiratory chain complex and OCR. When the antagonist of NMDARs was used before ischemic exposure, the neuronal mitochondrial dysfunction was alleviated, suggesting that these aberrant deviations of NMDARs from basal levels led to mitochondrial dysfunction. Furthermore, when the antagonist of NR2B was administrated intracerebroventricularly at the sub-acute cerebral ischemia, the volume of cerebral infarct region was decreased and the neural functions were improved. To sum up, the ratio of NR2B-containing NMDARs is vital for mitochondrial homeostasis and then neuronal survival. NR2B-targeted intervention should be chosen at the sub-acute stage of cerebral ischemia.
Collapse
Affiliation(s)
- Bei Guo
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Huimeng Song
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Jiahui Fan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Bin Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Lingyi Chen
- John Bapst Memorial High School, Bangor, CA, USA
| | - Qiandai Hu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Yanling Yin
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China.
| |
Collapse
|
5
|
Farzaei MH, Ramezani-Aliakbari F, Ramezani-Aliakbari M, Zarei M, Komaki A, Shahidi S, Sarihi A, Salehi I. Regulatory effects of trimetazidine in cardiac ischemia/reperfusion injury. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1633-1646. [PMID: 36971866 DOI: 10.1007/s00210-023-02469-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/19/2023] [Indexed: 03/29/2023]
Abstract
Ischemia/reperfusion (I/R) injury is a tissue damage during reperfusion after an ischemic condition. I/R injury is induced by pathological cases including stroke, myocardial infarction, circulatory arrest, sickle cell disease, acute kidney injury, trauma, and sleep apnea. It can lead to increased morbidity and mortality in the context of these processes. Mitochondrial dysfunction is one of the hallmarks of I/R insult, which is induced via reactive oxygen species (ROS) production, apoptosis, and autophagy. MicroRNAs (miRNAs, miRs) are non-coding RNAs that play a main regulatory role in gene expression. Recently, there are evidence, which miRNAs are the major modulators of cardiovascular diseases, especially myocardial I/R injury. Cardiovascular miRNAs, specifically miR-21, and probably miR-24 and miR-126 have protective effects on myocardial I/R injury. Trimetazidine (TMZ) is a new class of metabolic agents with an anti-ischemic activity. It has beneficial effects on chronic stable angina by suppressing mitochondrial permeability transition pore (mPTP) opening. The present review study addressed the different mechanistic effects of TMZ on cardiac I/R injury. Online databases including Scopus, PubMed, Web of Science, and Cochrane library were assessed for published studies between 1986 and 2021. TMZ, an antioxidant and metabolic agent, prevents the cardiac reperfusion injury by regulating AMP-activated protein kinase (AMPK), cystathionine-γ-lyase enzyme (CSE)/hydrogen sulfide (H2S), and miR-21. Therefore, TMZ protects the heart against I/R injury by inducing key regulators such as AMPK, CSE/H2S, and miR-21.
Collapse
Affiliation(s)
- Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Maryam Ramezani-Aliakbari
- Department of Medicinal Chemistry, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Zarei
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
6
|
Semenzato M, Kohr MJ, Quirin C, Menabò R, Alanova P, Alan L, Pellattiero A, Murphy E, Di Lisa F, Scorrano L. Oxidization of optic atrophy 1 cysteines occurs during heart ischemia-reperfusion and amplifies cell death by oxidative stress. Redox Biol 2023; 63:102755. [PMID: 37224696 DOI: 10.1016/j.redox.2023.102755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/01/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023] Open
Abstract
During cardiac ischemia-reperfusion, excess reactive oxygen species can damage mitochondrial, cellular and organ function. Here we show that cysteine oxidation of the mitochondrial protein Opa1 contributes to mitochondrial damage and cell death caused by oxidative stress. Oxy-proteomics of ischemic-reperfused hearts reveal oxidation of the C-terminal C786 of Opa1 and treatment of perfused mouse hearts, adult cardiomyocytes, and fibroblasts with H2O2 leads to the formation of a reduction-sensitive ∼180 KDa Opa1 complex, distinct from the ∼270 KDa one antagonizing cristae remodeling. This Opa1 oxidation process is curtailed by mutation of C786 and of the other 3 Cys residues of its C-terminal domain (Opa1TetraCys). When reintroduced in Opa1-/- cells, Opa1TetraCys is not efficiently processed into short Opa1TetraCys and hence fails to fuse mitochondria. Unexpectedly, Opa1TetraCys restores mitochondrial ultrastructure in Opa1-/- cells and protects them from H2O2-induced mitochondrial depolarization, cristae remodeling, cytochrome c release and cell death. Thus, preventing the Opa1 oxidation occurring during cardiac ischemia-reperfusion reduces mitochondrial damage and cell death induced by oxidative stress independent of mitochondrial fusion.
Collapse
Affiliation(s)
- Martina Semenzato
- Department of Biology, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padua, Italy
| | - Mark J Kohr
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Charlotte Quirin
- Department of Biology, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padua, Italy
| | - Roberta Menabò
- Department of Biomedical Sciences, University of Padova, Italy; National Research Council of Italy (CNR), Padova, Italy
| | - Petra Alanova
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Developmental Cardiology, Institute of Physiology CAS, Prague, Czech Republic
| | - Lukas Alan
- Department of Biology, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padua, Italy
| | - Anna Pellattiero
- Department of Biology, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padua, Italy
| | | | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Italy; National Research Council of Italy (CNR), Padova, Italy
| | - Luca Scorrano
- Department of Biology, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padua, Italy.
| |
Collapse
|
7
|
Russo I, Barale C, Melchionda E, Penna C, Pagliaro P. Platelets and Cardioprotection: The Role of Nitric Oxide and Carbon Oxide. Int J Mol Sci 2023; 24:ijms24076107. [PMID: 37047079 PMCID: PMC10094148 DOI: 10.3390/ijms24076107] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Nitric oxide (NO) and carbon monoxide (CO) represent a pair of biologically active gases with an increasingly well-defined range of effects on circulating platelets. These gases interact with platelets and cells in the vessels and heart and exert fundamentally similar biological effects, albeit through different mechanisms and with some peculiarity. Within the cardiovascular system, for example, the gases are predominantly vasodilators and exert antiaggregatory effects, and are protective against damage in myocardial ischemia-reperfusion injury. Indeed, NO is an important vasodilator acting on vascular smooth muscle and is able to inhibit platelet activation. NO reacts with superoxide anion (O2(-•)) to form peroxynitrite (ONOO(-)), a nitrosating agent capable of inducing oxidative/nitrative signaling and stress both at cardiovascular, platelet, and plasma levels. CO reduces platelet reactivity, therefore it is an anticoagulant, but it also has some cardioprotective and procoagulant properties. This review article summarizes current knowledge on the platelets and roles of gas mediators (NO, and CO) in cardioprotection. In particular, we aim to examine the link and interactions between platelets, NO, and CO and cardioprotective pathways.
Collapse
Affiliation(s)
- Isabella Russo
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Cristina Barale
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Elena Melchionda
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| |
Collapse
|
8
|
Burda R, Burda J, Morochovič R. Ischemic Tolerance—A Way to Reduce the Extent of Ischemia–Reperfusion Damage. Cells 2023; 12:cells12060884. [PMID: 36980225 PMCID: PMC10047660 DOI: 10.3390/cells12060884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023] Open
Abstract
Individual tissues have significantly different resistance to ischemia–reperfusion damage. There is still no adequate treatment for the consequences of ischemia–reperfusion damage. By utilizing ischemic tolerance, it is possible to achieve a significant reduction in the extent of the cell damage due to ischemia–reperfusion injury. Since ischemia–reperfusion damage usually occurs unexpectedly, the use of preconditioning is extremely limited. In contrast, postconditioning has wider possibilities for use in practice. In both cases, the activation of ischemic tolerance can also be achieved by the application of sublethal stress on a remote organ. Despite very encouraging and successful results in animal experiments, the clinical results have been disappointing so far. To avoid the factors that prevent the activation of ischemic tolerance, the solution has been to use blood plasma containing tolerance effectors. This plasma is taken from healthy donors in which, after exposure to two sublethal stresses within 48 h, effectors of ischemic tolerance occur in the plasma. Application of this activated plasma to recipient animals after the end of lethal ischemia prevents cell death and significantly reduces the consequences of ischemia–reperfusion damage. Until there is a clear chemical identification of the end products of ischemic tolerance, the simplest way of enhancing ischemic tolerance will be the preparation of activated plasma from young healthy donors with the possibility of its immediate use in recipients during the initial treatment.
Collapse
Affiliation(s)
- Rastislav Burda
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
- Correspondence:
| | - Jozef Burda
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Košice, Slovakia
| | - Radoslav Morochovič
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
| |
Collapse
|
9
|
Network-Assisted Systems Biology Analysis of the Mitochondrial Proteome in a Pre-Clinical Model of Ischemia, Revascularization and Post-Conditioning. Int J Mol Sci 2022; 23:ijms23042087. [PMID: 35216205 PMCID: PMC8879554 DOI: 10.3390/ijms23042087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/24/2022] Open
Abstract
Infarct size is the major risk predictor for developing heart failure after an acute myocardial infarction (AMI). The discovery of the conditioning phenomena (i.e., repetitive brief cycles of ischemia applied either before or after a prolonged ischemic insult) has highlighted the existence of endogenous protective mechanisms of the heart potentially limiting infarct size after revascularization. However, most cardioprotective strategies, aiming at infarct size reduction, have failed in clinical studies. Thus, cardioprotection is an unmet clinical need. In the present study, we took a network-assisted systems biology approach to explore the mitochondrial proteomic signature of the myocardium after ischemia, ischemia with direct revascularization, and ischemia with re-establishment of blood flow by post-conditioning in a swine model of AMI. Furthermore, network extension with the ENCODE project human regulatory data allowed the prediction of potential transcription factors at play in the response to post-conditioning of the myocardium. Collectively, our results identify cardiac metabolism as a driver of cardioprotection, highlighting a dual role for post-conditioning promoting metabolic reprogramming of the myocardium, and a protective response mediated by VDAC2 and DJ-1 in the mitochondria.
Collapse
|
10
|
Ischemia-Reperfusion Injury in Peripheral Artery Disease and Traditional Chinese Medicine Treatment. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4954070. [PMID: 34899949 PMCID: PMC8660193 DOI: 10.1155/2021/4954070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022]
Abstract
Peripheral artery disease (PAD) is a serious public health issue, characterized by circulation disorder of the lower extreme that reduces the physical activity of the lower extremity muscle. The artery narrowed by atherosclerotic lesions initiates limb ischemia. In the progression of treatment, reperfusion injury is still inevitable. Ischemia-reperfusion injury induced by PAD is responsible for hypoxia and nutrient deficiency. PAD triggers hindlimb ischemia and reperfusion (I/R) cycles through various mechanisms, mainly including mitochondrial dysfunction and inflammation. Alternatively, mitochondrial dysfunction plays a central role. The I/R injury may cause cells' injury and even death. However, the mechanism of I/R injury and the way of cell damage or death are still unclear. We review the pathophysiology of I/R injury, which is majorly about mitochondrial dysfunction. Then, we focus on the cell damage and death during I/R injury. Further comprehension of the progress of I/R will help identify biomarkers for diagnosis and therapeutic targets to PAD. In addition, traditional Chinese medicine has played an important role in the treatment of I/R injury, and we will make a brief introduction.
Collapse
|
11
|
Manousek J, Kala P, Lokaj P, Ondrus T, Helanova K, Miklikova M, Brazdil V, Tomandlova M, Parenica J, Pavkova Goldbergova M, Hlasensky J. Oxidative Stress in Takotsubo Syndrome-Is It Essential for an Acute Attack? Indirect Evidences Support Multisite Impact Including the Calcium Overload-Energy Failure Hypothesis. Front Cardiovasc Med 2021; 8:732708. [PMID: 34738019 PMCID: PMC8562109 DOI: 10.3389/fcvm.2021.732708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/16/2021] [Indexed: 12/28/2022] Open
Abstract
Indirect evidences in reviews and case reports on Takotsubo syndrome (TTS) support the fact that the existence of oxidative stress (OS) might be its common feature in the pre-acute stage. The sources of OS are exogenous (environmental factors including pharmacological and toxic influences) and endogenous, the combination of both may be present, and they are being discussed in detail. OS is associated with several pathological conditions representing TTS comorbidities and triggers. The dominant source of OS electrones are mitochondria. Our analysis of drug therapy related to acute TTS shows many interactions, e.g., cytostatics and glucocorticoids with mitochondrial cytochrome P450 and other enzymes important for OS. One of the most frequently discussed mechanisms in TTS is the effect of catecholamines on myocardium. Yet, their metabolic influence is neglected. OS is associated with the oxidation of catecholamines leading to the synthesis of their oxidized forms - aminochromes. Under pathological conditions, this pathway may dominate. There are evidences of interference between OS, catecholamine/aminochrome effects, their metabolism and antioxidant protection. The OS offensive may cause fast depletion of antioxidant protection including the homocystein-methionine system, whose activity decreases with age. The alteration of effector subcellular structures (mitochondria, sarco/endoplasmic reticulum) and subsequent changes in cellular energetics and calcium turnover may also occur and lead to the disruption of cellular function, including neurons and cardiomyocytes. On the organ level (nervous system and heart), neurocardiogenic stunning may occur. The effects of OS correspond to the effect of high doses of catecholamines in the experiment. Intensive OS might represent "conditio sine qua non" for this acute clinical condition. TTS might be significantly more complex pathology than currently perceived so far.
Collapse
Affiliation(s)
- Jan Manousek
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
| | - Petr Kala
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petr Lokaj
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Tomas Ondrus
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Katerina Helanova
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marie Miklikova
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
| | - Vojtech Brazdil
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marie Tomandlova
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jiri Parenica
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | | | - Jiri Hlasensky
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
12
|
McClung JA, Levy L, Garcia V, Stec DE, Peterson SJ, Abraham NG. Heme-oxygenase and lipid mediators in obesity and associated cardiometabolic diseases: Therapeutic implications. Pharmacol Ther 2021; 231:107975. [PMID: 34499923 DOI: 10.1016/j.pharmthera.2021.107975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity-mediated metabolic syndrome remains the leading cause of death worldwide. Among many potential targets for pharmacological intervention, a promising strategy involves the heme oxygenase (HO) system, specifically its inducible form, HO-1. This review collects and updates much of the current knowledge relevant to pharmacology and clinical medicine concerning HO-1 in metabolic diseases and its effect on lipid metabolism. HO-1 has pleotropic effects that collectively reduce inflammation, while increasing vasodilation and insulin and leptin sensitivity. Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Similarly, HO-1and bilirubin are potential therapeutic targets in the treatment of fat-induced liver diseases. HO-1-mediated upregulation of EET is capable not only of reversing endothelial dysfunction and hypertension, but also of reversing cardiac remodeling, a hallmark of the metabolic syndrome. This process involves browning of white fat tissue (i.e. formation of healthy adipocytes) and reduced lipotoxicity, which otherwise will be toxic to the heart. More importantly, this review examines the activity of EET in biological systems and a series of pathways that explain its mechanism of action and discusses how these might be exploited for potential therapeutic use. We also discuss the link between cardiac ectopic fat deposition and cardiac function in humans, which is similar to that described in obese mice and is regulated by HO-1-EET-PGC1α signaling, a potent negative regulator of the inflammatory adipokine NOV.
Collapse
Affiliation(s)
- John A McClung
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Lior Levy
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, United States of America.
| | - Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, United States of America; New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, United States of America
| | - Nader G Abraham
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America; Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America.
| |
Collapse
|
13
|
Uhlig C, Labus J. Volatile Versus Intravenous Anesthetics in Cardiac Anesthesia: a Narrative Review. CURRENT ANESTHESIOLOGY REPORTS 2021; 11:275-283. [PMID: 34276252 PMCID: PMC8271298 DOI: 10.1007/s40140-021-00466-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 11/25/2022]
Abstract
Purpose of the Review The present review addresses clinicians and gives an overview about the experimental rationale for pharmacological conditioning associated with volatile anesthetics, opioids, and propofol; the current clinical data; and the technical considerations regarding the clinical routine in cardiac anesthesia. Recent Findings Volatile anesthetics have been standard of care for general anesthesia for cardiac surgery, especially while using cardiopulmonary bypass. The 2019 published MYRIAD trial was not able to show a difference in mortality or cardiac biomarkers for volatile anesthetics compared to total intravenous anesthesia (TIVA), raising the question of equivalence with respect to patient outcome. Summary Reviewing the literature, the scientific foundation for the belief of clinically relevant conditioning by uninterrupted administration of a volatile anesthetic is weak. TIVA can also be performed safely in patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Christopher Uhlig
- Department of Cardiac Anesthesiology, Heart Center Dresden University Hospital, Fetscherstr. 76, 01307 Dresden, Germany
| | - Jakob Labus
- Department of Anesthesiology and Intensive Care Medicine, University Hospital and Medical Faculty, Cologne University, Albertus-Magnus-Platz, 50923 Cologne, Germany
| |
Collapse
|
14
|
Martins-Marques T, Rodriguez-Sinovas A, Girao H. Cellular crosstalk in cardioprotection: Where and when do reactive oxygen species play a role? Free Radic Biol Med 2021; 169:397-409. [PMID: 33892116 DOI: 10.1016/j.freeradbiomed.2021.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/14/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022]
Abstract
A well-balanced intercellular communication between the different cells within the heart is vital for the maintenance of cardiac homeostasis and function. Despite remarkable advances on disease management and treatment, acute myocardial infarction remains the major cause of morbidity and mortality worldwide. Gold standard reperfusion strategies, namely primary percutaneous coronary intervention, are crucial to preserve heart function. However, reestablishment of blood flow and oxygen levels to the infarcted area are also associated with an accumulation of reactive oxygen species (ROS), leading to oxidative damage and cardiomyocyte death, a phenomenon termed myocardial reperfusion injury. In addition, ROS signaling has been demonstrated to regulate multiple biological pathways, including cell differentiation and intercellular communication. Given the importance of cell-cell crosstalk in the coordinated response after cell injury, in this review, we will discuss the impact of ROS in the different forms of inter- and intracellular communication, as well as the role of gap junctions, tunneling nanotubes and extracellular vesicles in the propagation of oxidative damage in cardiac diseases, particularly in the context of ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Antonio Rodriguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall D'Hebron Institut de Recerca (VHIR), Vall D'Hebron Hospital Universitari, Vall D'Hebron Barcelona Hospital Campus, Passeig Vall D'Hebron, 119-129, 08035, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Henrique Girao
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
15
|
Panel M, Ahmed-Belkacem A, Ruiz I, Guichou JF, Pawlotsky JM, Ghaleh B, Morin D. A Phenyl-Pyrrolidine Derivative Reveals a Dual Inhibition Mechanism of Myocardial Mitochondrial Permeability Transition Pore, Which Is Limited by Its Myocardial Distribution. J Pharmacol Exp Ther 2021; 376:348-357. [PMID: 33303698 DOI: 10.1124/jpet.120.000359] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/08/2020] [Indexed: 03/08/2025] Open
Abstract
Mitochondrial permeability transition pore (mPTP) opening is a key event in cell death during myocardial ischemia reperfusion. Inhibition of its modulator cyclophilin D (CypD) by cyclosporine A (CsA) reduces ischemia-reperfusion injury. The use of cyclosporine A in this indication is debated; however, targeting mPTP remains a major goal to achieve. We investigated the protective effects of a new original small-molecule cyclophilin inhibitor C31, which was specifically designed to target CypD. CypD peptidylprolyl cis-trans isomerase (PPIase) activity was assessed by the standard chemotrypsin-coupled assay. The effects of C31 on mPTP opening were investigated in isolated mouse cardiac mitochondria by measuring mitochondrial swelling and calcium retention capacity (CRC) in rat H9C2 cardiomyoblasts and in adult mouse cardiomyocytes by fluorescence microscopy in isolated perfused mouse hearts and ex vivo after drug infusion in mice. C31 potently inhibited CypD PPIase activity and mitochondrial swelling. C31 was more effective at increasing mitochondrial CRC than CsA and was still able to increase CRC in Ppif -/- (CypD-inactivated) cardiac mitochondria. C31 delayed both mPTP opening and cell death in cardiomyocytes subjected to hypoxia reoxygenation. However, high concentrations of both drugs were necessary to reduce mPTP opening in isolated perfused hearts, and neither CsA nor C31 inhibited mPTP opening in heart after in vivo infusion, underlying the importance of myocardial drug distribution for cardioprotection. C31 is an original inhibitor of mPTP opening involving both CypD-dependent and -independent mechanisms. It constitutes a promising new cytoprotective agent. Optimization of its pharmacokinetic properties is now required prior to its use against cardiac ischemia-reperfusion injury. SIGNIFICANCE STATEMENT: This study demonstrates that the new cyclophilin inhibitor C31 potently inhibits cardiac mitochondrial permeability transition pore (mPTP) opening in vitro and ex vivo. The dual mechanism of action of C31 allows the prevention of mPTP opening beyond cyclophilin D inhibition. Further development of the compound might bring promising drug candidates for cardioprotection. However, the lack of effect of both C31 and cyclosporine A after systemic administration demonstrates the difficulties of targeting myocardial mitochondria in vivo and should be taken into account in cardioprotective strategies.
Collapse
Affiliation(s)
- Mathieu Panel
- U955 - IMRB, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France (M.P., B.G., D.M.);Université Paris-Est, UMR S955, UPEC, Créteil, France (A.A.B., I.R., J.M.P.); INSERM U955, Team « Viruses, Hepatology, Cancer », Hôpital Henri Mondor, Créteil, France (A.A.B., J.M.P., I.R.); Centre de Biochimie Structurale (CBS), INSERM U1054, CNRS UMR5048, Université de Montpellier, Montpellier, France (J.F.G.); and Department of Virology, Hôpital Henri Mondor, Créteil, France (J.M.P.)
| | - Abdelhakim Ahmed-Belkacem
- U955 - IMRB, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France (M.P., B.G., D.M.);Université Paris-Est, UMR S955, UPEC, Créteil, France (A.A.B., I.R., J.M.P.); INSERM U955, Team « Viruses, Hepatology, Cancer », Hôpital Henri Mondor, Créteil, France (A.A.B., J.M.P., I.R.); Centre de Biochimie Structurale (CBS), INSERM U1054, CNRS UMR5048, Université de Montpellier, Montpellier, France (J.F.G.); and Department of Virology, Hôpital Henri Mondor, Créteil, France (J.M.P.)
| | - Isaac Ruiz
- U955 - IMRB, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France (M.P., B.G., D.M.);Université Paris-Est, UMR S955, UPEC, Créteil, France (A.A.B., I.R., J.M.P.); INSERM U955, Team « Viruses, Hepatology, Cancer », Hôpital Henri Mondor, Créteil, France (A.A.B., J.M.P., I.R.); Centre de Biochimie Structurale (CBS), INSERM U1054, CNRS UMR5048, Université de Montpellier, Montpellier, France (J.F.G.); and Department of Virology, Hôpital Henri Mondor, Créteil, France (J.M.P.)
| | - Jean-François Guichou
- U955 - IMRB, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France (M.P., B.G., D.M.);Université Paris-Est, UMR S955, UPEC, Créteil, France (A.A.B., I.R., J.M.P.); INSERM U955, Team « Viruses, Hepatology, Cancer », Hôpital Henri Mondor, Créteil, France (A.A.B., J.M.P., I.R.); Centre de Biochimie Structurale (CBS), INSERM U1054, CNRS UMR5048, Université de Montpellier, Montpellier, France (J.F.G.); and Department of Virology, Hôpital Henri Mondor, Créteil, France (J.M.P.)
| | - Jean-Michel Pawlotsky
- U955 - IMRB, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France (M.P., B.G., D.M.);Université Paris-Est, UMR S955, UPEC, Créteil, France (A.A.B., I.R., J.M.P.); INSERM U955, Team « Viruses, Hepatology, Cancer », Hôpital Henri Mondor, Créteil, France (A.A.B., J.M.P., I.R.); Centre de Biochimie Structurale (CBS), INSERM U1054, CNRS UMR5048, Université de Montpellier, Montpellier, France (J.F.G.); and Department of Virology, Hôpital Henri Mondor, Créteil, France (J.M.P.)
| | - Bijan Ghaleh
- U955 - IMRB, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France (M.P., B.G., D.M.);Université Paris-Est, UMR S955, UPEC, Créteil, France (A.A.B., I.R., J.M.P.); INSERM U955, Team « Viruses, Hepatology, Cancer », Hôpital Henri Mondor, Créteil, France (A.A.B., J.M.P., I.R.); Centre de Biochimie Structurale (CBS), INSERM U1054, CNRS UMR5048, Université de Montpellier, Montpellier, France (J.F.G.); and Department of Virology, Hôpital Henri Mondor, Créteil, France (J.M.P.)
| | - Didier Morin
- U955 - IMRB, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France (M.P., B.G., D.M.);Université Paris-Est, UMR S955, UPEC, Créteil, France (A.A.B., I.R., J.M.P.); INSERM U955, Team « Viruses, Hepatology, Cancer », Hôpital Henri Mondor, Créteil, France (A.A.B., J.M.P., I.R.); Centre de Biochimie Structurale (CBS), INSERM U1054, CNRS UMR5048, Université de Montpellier, Montpellier, France (J.F.G.); and Department of Virology, Hôpital Henri Mondor, Créteil, France (J.M.P.)
| |
Collapse
|
16
|
Andreadou I, Schulz R, Papapetropoulos A, Turan B, Ytrehus K, Ferdinandy P, Daiber A, Di Lisa F. The role of mitochondrial reactive oxygen species, NO and H 2 S in ischaemia/reperfusion injury and cardioprotection. J Cell Mol Med 2020; 24:6510-6522. [PMID: 32383522 PMCID: PMC7299678 DOI: 10.1111/jcmm.15279] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/04/2020] [Accepted: 03/08/2020] [Indexed: 12/12/2022] Open
Abstract
Redox signalling in mitochondria plays an important role in myocardial ischaemia/reperfusion (I/R) injury and in cardioprotection. Reactive oxygen and nitrogen species (ROS/RNS) modify cellular structures and functions by means of covalent changes in proteins including among others S‐nitros(yl)ation by nitric oxide (NO) and its derivatives, and S‐sulphydration by hydrogen sulphide (H2S). Many enzymes are involved in the mitochondrial formation and handling of ROS, NO and H2S under physiological and pathological conditions. In particular, the balance between formation and removal of reactive species is impaired during I/R favouring their accumulation. Therefore, various interventions aimed at decreasing mitochondrial ROS accumulation have been developed and have shown cardioprotective effects in experimental settings. However, ROS, NO and H2S play also a role in endogenous cardioprotection, as in the case of ischaemic pre‐conditioning, so that preventing their increase might hamper self‐defence mechanisms. The aim of the present review was to provide a critical analysis of formation and role of reactive species, NO and H2S in mitochondria, with a special emphasis on mechanisms of injury and protection that determine the fate of hearts subjected to I/R. The elucidation of the signalling pathways of ROS, NO and H2S is likely to reveal novel molecular targets for cardioprotection that could be modulated by pharmacological agents to prevent I/R injury.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Andreas Daiber
- Molecular Cardiology, Center for Cardiology 1, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Fabio Di Lisa
- Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| |
Collapse
|
17
|
Antonucci S, Di Sante M, Sileikyte J, Deveraux J, Bauer T, Bround MJ, Menabò R, Paillard M, Alanova P, Carraro M, Ovize M, Molkentin JD, Cohen M, Forte MA, Bernardi P, Di Lisa F, Murphy E. A novel class of cardioprotective small-molecule PTP inhibitors. Pharmacol Res 2019; 151:104548. [PMID: 31759087 DOI: 10.1016/j.phrs.2019.104548] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/03/2023]
Abstract
Ischemia/reperfusion (I/R) injury is mediated in large part by opening of the mitochondrial permeability transition pore (PTP). Consequently, inhibitors of the PTP hold great promise for the treatment of a variety of cardiovascular disorders. At present, PTP inhibition is obtained only through the use of drugs (e.g. cyclosporine A, CsA) targeting cyclophilin D (CyPD) which is a key modulator, but not a structural component of the PTP. This limitation might explain controversial findings in clinical studies. Therefore, we investigated the protective effects against I/R injury of small-molecule inhibitors of the PTP (63 and TR002) that do not target CyPD. Both compounds exhibited a dose-dependent inhibition of PTP opening in isolated mitochondria and were more potent than CsA. Notably, PTP inhibition was observed also in mitochondria devoid of CyPD. Compounds 63 and TR002 prevented PTP opening and mitochondrial depolarization induced by Ca2+ overload and by reactive oxygen species in neonatal rat ventricular myocytes (NRVMs). Remarkably, both compounds prevented cell death, contractile dysfunction and sarcomeric derangement induced by anoxia/reoxygenation injury in NRVMs at sub-micromolar concentrations, and were more potent than CsA. Cardioprotection was observed also in adult mouse ventricular myocytes and human iPSc-derived cardiomyocytes, as well as ex vivo in perfused hearts. Thus, this study demonstrates that 63 and TR002 represent novel cardioprotective agents that inhibit PTP opening independent of CyPD targeting.
Collapse
Affiliation(s)
| | - Moises Di Sante
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Justina Sileikyte
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Jordan Deveraux
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Tyler Bauer
- Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Michael J Bround
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Roberta Menabò
- Department of Biomedical Sciences, University of Padova, Padova, Italy; National Research Council of Italy (CNR), Padova, Italy
| | - Melanie Paillard
- CarMeN Laboratory, University Claude Bernard Lyon 1, INSA Lyon, Oullins, France
| | - Petra Alanova
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Developmental Cardiology, Institute of Physiology CAS, Prague, Czech Republic
| | - Michela Carraro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Michel Ovize
- CarMeN Laboratory, University Claude Bernard Lyon 1, INSA Lyon, Oullins, France
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA; Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael Cohen
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Michael A Forte
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy; National Research Council of Italy (CNR), Padova, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova, Italy; National Research Council of Italy (CNR), Padova, Italy.
| | | |
Collapse
|
18
|
Antonucci S, Mulvey JF, Burger N, Di Sante M, Hall AR, Hinchy EC, Caldwell ST, Gruszczyk AV, Deshwal S, Hartley RC, Kaludercic N, Murphy MP, Di Lisa F, Krieg T. Selective mitochondrial superoxide generation in vivo is cardioprotective through hormesis. Free Radic Biol Med 2019; 134:678-687. [PMID: 30731114 PMCID: PMC6607027 DOI: 10.1016/j.freeradbiomed.2019.01.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 01/14/2023]
Abstract
Reactive oxygen species (ROS) have an equivocal role in myocardial ischaemia reperfusion injury. Within the cardiomyocyte, mitochondria are both a major source and target of ROS. We evaluate the effects of a selective, dose-dependent increase in mitochondrial ROS levels on cardiac physiology using the mitochondria-targeted redox cycler MitoParaquat (MitoPQ). Low levels of ROS decrease the susceptibility of neonatal rat ventricular myocytes (NRVMs) to anoxia/reoxygenation injury and also cause profound protection in an in vivo mouse model of ischaemia/reperfusion. However higher doses of MitoPQ resulted in a progressive alteration of intracellular [Ca2+] homeostasis and mitochondrial function in vitro, leading to dysfunction and death at high doses. Our data show that a primary increase in mitochondrial ROS can alter cellular function, and support a hormetic model in which low levels of ROS are cardioprotective while higher levels of ROS are cardiotoxic.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis
- Disease Models, Animal
- Herbicides/pharmacology
- Hormesis
- Male
- Mice
- Mice, Inbred C57BL
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Paraquat/pharmacology
- Rats
- Rats, Wistar
- Superoxides/metabolism
Collapse
Affiliation(s)
- Salvatore Antonucci
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - John F Mulvey
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Nils Burger
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Moises Di Sante
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Andrew R Hall
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Elizabeth C Hinchy
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | | | - Anja V Gruszczyk
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Soni Deshwal
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | | | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), 35131, Padova, Italy
| | - Michael P Murphy
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy; Neuroscience Institute, National Research Council of Italy (CNR), 35131, Padova, Italy.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
19
|
Aghaei M, Motallebnezhad M, Ghorghanlu S, Jabbari A, Enayati A, Rajaei M, Pourabouk M, Moradi A, Alizadeh AM, Khori V. Targeting autophagy in cardiac ischemia/reperfusion injury: A novel therapeutic strategy. J Cell Physiol 2019; 234:16768-16778. [PMID: 30807647 DOI: 10.1002/jcp.28345] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 12/31/2022]
Abstract
Acute myocardial infarction (AMI) is one of the leading causes of morbidity worldwide. Myocardial reperfusion is known as an effective therapeutic choice against AMI. However, reperfusion of blood flow induces ischemia/reperfusion (I/R) injury through different complex processes including ion accumulation, disruption of mitochondrial membrane potential, the formation of reactive oxygen species, and so forth. One of the processes that gets activated in response to I/R injury is autophagy. Indeed, autophagy acts as a "double-edged sword" in the pathology of myocardial I/R injury and there is a controversy about autophagy being beneficial or detrimental. On the basis of the autophagy effect and regulation on myocardial I/R injury, many studies targeted it as a therapeutic strategy. In this review, we discuss the role of autophagy in I/R injury and its targeting as a therapeutic strategy.
Collapse
Affiliation(s)
- Mehrdad Aghaei
- Rheumatology Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Morteza Motallebnezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Ghorghanlu
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Jabbari
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Pharmacognosy, Faculty of Pharmacy and Medicinal Plants Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Rajaei
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mona Pourabouk
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alireza Moradi
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
20
|
Xia Z, Li H, Irwin MG. Myocardial ischaemia reperfusion injury: the challenge of translating ischaemic and anaesthetic protection from animal models to humans. Br J Anaesth 2018; 117 Suppl 2:ii44-ii62. [PMID: 27566808 DOI: 10.1093/bja/aew267] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myocardial ischaemia reperfusion injury is the leading cause of death in patients with cardiovascular disease. Interventions such as ischaemic pre and postconditioning protect against myocardial ischaemia reperfusion injury. Certain anaesthesia drugs and opioids can produce the same effects, which led to an initial flurry of excitement given the extensive use of these drugs in surgery. The underlying mechanisms have since been extensively studied in experimental animal models but attempts to translate these findings to clinical settings have resulted in contradictory results. There are a number of reasons for this such as dose response, the intensity of the ischaemic stimulus applied, the duration of ischaemia and lost or diminished cardioprotection in common co-morbidities such as diabetes and senescence. This review focuses on current knowledge regarding myocardial ischaemia reperfusion injury and cardioprotective interventions both in experimental animal studies and in clinical trials.
Collapse
Affiliation(s)
- Z Xia
- Department of Anaesthesiology Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong SAR, China
| | - H Li
- Department of Anaesthesiology
| | - M G Irwin
- Department of Anaesthesiology Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Hentia C, Rizzato A, Camporesi E, Yang Z, Muntean DM, Săndesc D, Bosco G. An overview of protective strategies against ischemia/reperfusion injury: The role of hyperbaric oxygen preconditioning. Brain Behav 2018; 8:e00959. [PMID: 29761012 PMCID: PMC5943756 DOI: 10.1002/brb3.959] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 02/12/2018] [Accepted: 02/18/2018] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Ischemia/reperfusion (I/R) injury, such as myocardial infarction, stroke, and peripheral vascular disease, has been recognized as the most frequent causes of devastating disorders and death currently. Protective effect of various preconditioning stimuli, including hyperbaric oxygen (HBO), has been proposed in the management of I/R. METHODS In this study, we searched and reviewed up-to-date published papers to explore the pathophysiology of I/R injury and to understand the mechanisms underlying the protective effect of HBO as conditioning strategy. RESULTS Animal study and clinic observation support the notion that HBO therapy and conditioning provide beneficial effect against the deleterious effects of postischemic reperfusion. Several explanations have been proposed. The first likely mechanism may be that HBO counteracts hypoxia and reduces I/R injury by improving oxygen delivery to an area with diminished blood flow. Secondly, by reducing hypoxia-ischemia, HBO reduces all the pathological events as a consequence of hypoxia, including tissue edema, increased affective area permeability, postischemia derangement of tissue metabolism, and inflammation. Thirdly, HBO may directly affect cell apoptosis, signal transduction, and gene expression in those that are sensitive to oxygen or hypoxia. HBO provides a reservoir of oxygen at cellular level not only carried by blood, but also by diffusion from the interstitial tissue where it reaches high concentration that may last for several hours, improves endothelial function and rheology, and decreases local inflammation and edema. CONCLUSION Evidence suggests the benefits of HBO when used as a preconditioning stimulus in the setting of I/R injury. Translating the beneficial effects of HBO into current practice requires, as for the "conditioning strategies", a thorough consideration of risk factors, comorbidities, and comedications that could interfere with HBO-related protection.
Collapse
Affiliation(s)
- Ciprian Hentia
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy.,Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Alex Rizzato
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy
| | | | - Zhongjin Yang
- The Institute for Human Performance SUNY Upstate Medical University Syracuse NY USA
| | - Danina M Muntean
- Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania.,Center for Translational Research and Systems Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Dorel Săndesc
- Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Gerardo Bosco
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy
| |
Collapse
|
22
|
Pagliaro P, Femminò S, Popara J, Penna C. Mitochondria in Cardiac Postconditioning. Front Physiol 2018; 9:287. [PMID: 29632499 PMCID: PMC5879113 DOI: 10.3389/fphys.2018.00287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a pivotal role in cardioprotection. Here we report some fundamental studies which considered the role of mitochondrial components (connexin 43, mitochondrial KATP channels and mitochondrial permeability transition pore) in postconditioning cardioprotection. We briefly discuss the role of mitochondria, reactive oxygen species and gaseous molecules in postconditioning. Also the effects of anesthetics-used as cardioprotective substances-is briefly considered in the context of postconditioning. The role of mitochondrial postconditioning signaling in determining the limitation of cell death is underpinned. Issues in clinical translation are briefly considered. The aim of the present mini-review is to discuss in a historical perspective the role of main mitochondria mechanisms in cardiac postconditioning.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Jasmin Popara
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
23
|
Parra V, Altamirano F, Hernández-Fuentes CP, Tong D, Kyrychenko V, Rotter D, Pedrozo Z, Hill JA, Eisner V, Lavandero S, Schneider JW, Rothermel BA. Down Syndrome Critical Region 1 Gene, Rcan1, Helps Maintain a More Fused Mitochondrial Network. Circ Res 2018; 122:e20-e33. [PMID: 29362227 DOI: 10.1161/circresaha.117.311522] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 01/05/2023]
Abstract
RATIONALE The regulator of calcineurin 1 (RCAN1) inhibits CN (calcineurin), a Ca2+-activated protein phosphatase important in cardiac remodeling. In humans, RCAN1 is located on chromosome 21 in proximity to the Down syndrome critical region. The hearts and brains of Rcan1 KO mice are more susceptible to damage from ischemia/reperfusion (I/R); however, the underlying cause is not known. OBJECTIVE Mitochondria are key mediators of I/R damage. The goal of these studies was to determine the impact of RCAN1 on mitochondrial dynamics and function. METHODS AND RESULTS Using both neonatal and isolated adult cardiomyocytes, we show that, when RCAN1 is depleted, the mitochondrial network is more fragmented because of increased CN-dependent activation of the fission protein, DRP1 (dynamin-1-like). Mitochondria in RCAN1-depleted cardiomyocytes have reduced membrane potential, O2 consumption, and generation of reactive oxygen species, as well as a reduced capacity for mitochondrial Ca2+ uptake. RCAN1-depleted cardiomyocytes were more sensitive to I/R; however, pharmacological inhibition of CN, DRP1, or CAPN (calpains; Ca2+-activated proteases) restored protection, suggesting that in the absence of RCAN1, CAPN-mediated damage after I/R is greater because of a decrease in the capacity of mitochondria to buffer cytoplasmic Ca2+. Increasing RCAN1 levels by adenoviral infection was sufficient to enhance fusion and confer protection from I/R. To examine the impact of more modest, and biologically relevant, increases in RCAN1, we compared the mitochondrial network in induced pluripotent stem cells derived from individuals with Down syndrome to that of isogenic, disomic controls. Mitochondria were more fused, and O2 consumption was greater in the trisomic induced pluripotent stem cells; however, coupling efficiency and metabolic flexibility were compromised compared with disomic induced pluripotent stem cells. Depletion of RCAN1 from trisomic induced pluripotent stem cells was sufficient to normalize mitochondrial dynamics and function. CONCLUSIONS RCAN1 helps maintain a more interconnected mitochondrial network, and maintaining appropriate RCAN1 levels is important to human health and disease.
Collapse
Affiliation(s)
- Valentina Parra
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.).
| | - Francisco Altamirano
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Carolina P Hernández-Fuentes
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Dan Tong
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Victoriia Kyrychenko
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - David Rotter
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Zully Pedrozo
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Joseph A Hill
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Verónica Eisner
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Sergio Lavandero
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Jay W Schneider
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Beverly A Rothermel
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.).
| |
Collapse
|
24
|
Ivanina AV, Nesmelova I, Leamy L, Sokolov EP, Sokolova IM. Intermittent hypoxia leads to functional reorganization of mitochondria and affects cellular bioenergetics in marine molluscs. ACTA ACUST UNITED AC 2017; 219:1659-74. [PMID: 27252455 DOI: 10.1242/jeb.134700] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 03/14/2016] [Indexed: 12/11/2022]
Abstract
Fluctuations in oxygen (O2) concentrations represent a major challenge to aerobic organisms and can be extremely damaging to their mitochondria. Marine intertidal molluscs are well-adapted to frequent O2 fluctuations, yet it remains unknown how their mitochondrial functions are regulated to sustain energy metabolism and prevent cellular damage during hypoxia and reoxygenation (H/R). We used metabolic control analysis to investigate the mechanisms of mitochondrial responses to H/R stress (18 h at <0.1% O2 followed by 1 h of reoxygenation) using hypoxia-tolerant intertidal clams Mercenaria mercenaria and hypoxia-sensitive subtidal scallops Argopecten irradians as models. We also assessed H/R-induced changes in cellular energy balance, oxidative damage and unfolded protein response to determine the potential links between mitochondrial dysfunction and cellular injury. Mitochondrial responses to H/R in scallops strongly resembled those in other hypoxia-sensitive organisms. Exposure to hypoxia followed by reoxygenation led to a strong decrease in the substrate oxidation (SOX) and phosphorylation (PHOS) capacities as well as partial depolarization of mitochondria of scallops. Elevated mRNA expression of a reactive oxygen species-sensitive enzyme aconitase and Lon protease (responsible for degradation of oxidized mitochondrial proteins) during H/R stress was consistent with elevated levels of oxidative stress in mitochondria of scallops. In hypoxia-tolerant clams, mitochondrial SOX capacity was enhanced during hypoxia and continued rising during the first hour of reoxygenation. In both species, the mitochondrial PHOS capacity was suppressed during hypoxia, likely to prevent ATP wastage by the reverse action of FO,F1-ATPase. The PHOS capacity recovered after 1 h of reoxygenation in clams but not in scallops. Compared with scallops, clams showed a greater suppression of energy-consuming processes (such as protein turnover and ion transport) during hypoxia, indicated by inactivation of the translation initiation factor EIF-2α, suppression of 26S proteasome activity and a dramatic decrease in the activity of Na(+)/K(+)-ATPase. The steady-state levels of adenylates were preserved during H/R exposure and AMP-dependent protein kinase was not activated in either species, indicating that the H/R exposure did not lead to severe energy deficiency. Taken together, our findings suggest that mitochondrial reorganizations sustaining high oxidative phosphorylation flux during recovery, combined with the ability to suppress ATP-demanding cellular functions during hypoxia, may contribute to high resilience of clams to H/R stress and help maintain energy homeostasis during frequent H/R cycles in the intertidal zone.
Collapse
Affiliation(s)
- Anna V Ivanina
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Irina Nesmelova
- Department of Physics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Larry Leamy
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Eugene P Sokolov
- Department of General Surgery, Carolinas Medical Center, Charlotte, NC 28232, USA
| | - Inna M Sokolova
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
25
|
Steven S, Daiber A, Dopheide JF, Münzel T, Espinola-Klein C. Peripheral artery disease, redox signaling, oxidative stress - Basic and clinical aspects. Redox Biol 2017; 12:787-797. [PMID: 28437655 PMCID: PMC5403804 DOI: 10.1016/j.redox.2017.04.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/28/2017] [Accepted: 04/10/2017] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen and nitrogen species (ROS and RNS, e.g. H2O2, nitric oxide) confer redox regulation of essential cellular signaling pathways such as cell differentiation, proliferation, migration and apoptosis. At higher concentrations, ROS and RNS lead to oxidative stress and oxidative damage of biomolecules (e.g. via formation of peroxynitrite, fenton chemistry). Peripheral artery disease (PAD) is characterized by severe ischemic conditions in the periphery leading to intermittent claudication and critical limb ischemia (end stage). It is well known that redox biology and oxidative stress play an important role in this setting. We here discuss the major pathways of oxidative stress and redox signaling underlying the disease progression with special emphasis on the contribution of inflammatory processes. We also highlight therapeutic strategies comprising pharmacological (e.g. statins, angiotensin-converting enzyme inhibitors, phosphodiesterase inhibition) and non-pharmacological (e.g. exercise) interventions. Both of these strategies induce potent indirect antioxidant and anti-inflammatory mechanisms that may contribute to an improvement of PAD associated complications and disease progression by removing excess formation of ROS and RNS (e.g. by ameliorating primary complications such as hyperlipidemia and hypertension) as well as the normalization of the inflammatory phenotype suppressing the progression of atherosclerosis.
Collapse
Affiliation(s)
- Sebastian Steven
- Laboratory of Molecular Cardiology, Center of Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Daiber
- Laboratory of Molecular Cardiology, Center of Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Jörn F Dopheide
- Angiology, Center of Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany; Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas Münzel
- Laboratory of Molecular Cardiology, Center of Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christine Espinola-Klein
- Angiology, Center of Cardiology, Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
26
|
Abstract
The heart of patients with hypertension and cardiac hypertrophy is more vulnerable to ischemia-reperfusion injury (IRI). Here we discuss the main mechanisms of IRI and possible targets for cardioprotection. In particular, we consider the viewpoint that hypertension and cardiac hypertrophy may act synergistically in increasing the predisposition to cardiovascular accidents and in worsening IRI. There is no doubt that hypertrophic hearts may be redirected to be less vulnerable to IRI. Some experimental evidences suggest that antihypertensive drugs may have beneficial effects, some of which are not directly related to hypertension-lowering effect. However, more thorough experimental and clinical studies are necessary to understand the mechanisms and to maximize the beneficial effects of reperfusion after a heart attack in the presence of comorbidities, such as hypertension and cardiac hypertrophy.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | | |
Collapse
|
27
|
Daiber A, Di Lisa F, Oelze M, Kröller‐Schön S, Steven S, Schulz E, Münzel T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol 2017; 174:1670-1689. [PMID: 26660451 PMCID: PMC5446573 DOI: 10.1111/bph.13403] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are associated with and/or caused by oxidative stress. This concept has been proven by using the approach of genetic deletion of reactive species producing (pro-oxidant) enzymes as well as by the overexpression of reactive species detoxifying (antioxidant) enzymes leading to a marked reduction of reactive oxygen and nitrogen species (RONS) and in parallel to an amelioration of the severity of diseases. Likewise, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of antioxidant RONS detoxifying enzymes. Thus, the consequences of the interaction (redox crosstalk) of superoxide/hydrogen peroxide produced by mitochondria with other ROS producing enzymes such as NADPH oxidases (Nox) are of outstanding importance and will be discussed including the consequences for endothelial nitric oxide synthase (eNOS) uncoupling as well as the redox regulation of the vascular function/tone in general (soluble guanylyl cyclase, endothelin-1, prostanoid synthesis). Pathways and potential mechanisms leading to this crosstalk will be analysed in detail and highlighted by selected examples from the current literature including hypoxia, angiotensin II-induced hypertension, nitrate tolerance, aging and others. The general concept of redox-based activation of RONS sources via "kindling radicals" and enzyme-specific "redox switches" will be discussed providing evidence that mitochondria represent key players and amplifiers of the burden of oxidative stress. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Fabio Di Lisa
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Matthias Oelze
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Swenja Kröller‐Schön
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Sebastian Steven
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- Center of Thrombosis and HemostasisMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Eberhard Schulz
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Thomas Münzel
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| |
Collapse
|
28
|
McDougal AD, Dewey CF. Modeling oxygen requirements in ischemic cardiomyocytes. J Biol Chem 2017; 292:11760-11776. [PMID: 28487363 DOI: 10.1074/jbc.m116.751826] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 05/01/2017] [Indexed: 11/06/2022] Open
Abstract
Heart disease remains the leading cause of death globally. Although reperfusion following myocardial ischemia can prevent death by restoring nutrient flow, ischemia/reperfusion injury can cause significant heart damage. The mechanisms that drive ischemia/reperfusion injury are not well understood; currently, few methods can predict the state of the cardiac muscle cell and its metabolic conditions during ischemia. Here, we explored the energetic sustainability of cardiomyocytes, using a model for cellular metabolism to predict the levels of ATP following hypoxia. We modeled glycolytic metabolism with a system of coupled ordinary differential equations describing the individual metabolic reactions within the cardiomyocyte over time. Reduced oxygen levels and ATP consumption rates were simulated to characterize metabolite responses to ischemia. By tracking biochemical species within the cell, our model enables prediction of the cell's condition up to the moment of reperfusion. The simulations revealed a distinct transition between energetically sustainable and unsustainable ATP concentrations for various energetic demands. Our model illustrates how even low oxygen concentrations allow the cell to perform essential functions. We found that the oxygen level required for a sustainable level of ATP increases roughly linearly with the ATP consumption rate. An extracellular O2 concentration of ∼0.007 mm could supply basic energy needs in non-beating cardiomyocytes, suggesting that increased collateral circulation may provide an important source of oxygen to sustain the cardiomyocyte during extended ischemia. Our model provides a time-dependent framework for studying various intervention strategies to change the outcome of reperfusion.
Collapse
Affiliation(s)
- Anthony D McDougal
- Departments of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - C Forbes Dewey
- Departments of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.
| |
Collapse
|
29
|
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: 25] [Impact Index Per Article: 3.1] [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
|
30
|
Dhivya V, Priya LB, Chirayil HT, Sathiskumar S, Huang CY, Padma VV. Piperine modulates isoproterenol induced myocardial ischemia through antioxidant and anti-dyslipidemic effect in male Wistar rats. Biomed Pharmacother 2017; 87:705-713. [DOI: 10.1016/j.biopha.2017.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/19/2016] [Accepted: 01/01/2017] [Indexed: 12/30/2022] Open
|
31
|
Delay C, Paradis S, Charles AL, Thaveau F, Chenesseau B, Zoll J, Chakfe N, Geny B, Lejay A. [Skeletal muscle ischemia-reperfusion and ischemic conditioning pathophysiology-clinical applications for the vascular surgeon]. JOURNAL DE MEDECINE VASCULAIRE 2017; 42:29-38. [PMID: 27989659 DOI: 10.1016/j.jmv.2016.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/15/2016] [Indexed: 06/06/2023]
Abstract
Ischemia-reperfusion, which is characterized by deficient oxygen supply and subsequent restoration of blood flow, can cause irreversible damage to tissue. The vascular surgeon is daily faced with ischemia-reperfusion situations. Indeed, arterial clamping induces ischemia, followed by reperfusion when declamping. Mechanisms underlying ischemia-reperfusion injury are complex and multifactorial. Increases in cellular calcium and reactive oxygen species, initiated during ischemia and then amplified upon reperfusion are thought to be the main mediators of reperfusion injury. Mitochondrial dysfunction also plays an important role. Extensive research has focused on increasing skeletal muscle tolerance to ischemia-reperfusion injury, especially through the use of ischemic conditioning strategies. The purpose of this review is to focus on the cellular responses associated with ischemia-reperfusion, as well as to discuss the effects of ischemic conditioning strategies. This would help the vascular surgeon in daily practice, in order to try to improve surgical outcome in the setting of ischemia-reperfusion.
Collapse
Affiliation(s)
- C Delay
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France
| | - S Paradis
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - A L Charles
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - F Thaveau
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France
| | - B Chenesseau
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France
| | - J Zoll
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - N Chakfe
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France; Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - B Geny
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - A Lejay
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France; Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France.
| |
Collapse
|
32
|
Bagheri F, Khori V, Alizadeh AM, Khalighfard S, Khodayari S, Khodayari H. Reactive oxygen species-mediated cardiac-reperfusion injury: Mechanisms and therapies. Life Sci 2016; 165:43-55. [DOI: 10.1016/j.lfs.2016.09.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/13/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022]
|
33
|
Epoxyeicosatrienoic Acid as Therapy for Diabetic and Ischemic Cardiomyopathy. Trends Pharmacol Sci 2016; 37:945-962. [DOI: 10.1016/j.tips.2016.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/12/2016] [Accepted: 08/17/2016] [Indexed: 12/19/2022]
|
34
|
Onukwufor JO, Kibenge F, Stevens D, Kamunde C. Hypoxia-reoxygenation differentially alters the thermal sensitivity of complex I basal and maximal mitochondrial oxidative capacity. Comp Biochem Physiol A Mol Integr Physiol 2016; 201:87-94. [DOI: 10.1016/j.cbpa.2016.06.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/17/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
|
35
|
Witowski NE, Lusczek ER, Determan CE, Lexcen DR, Mulier KE, Wolf A, Ostrowski BG, Beilman GJ. Metabolomic analysis of survival in carbohydrate pre-fed pigs subjected to shock and polytrauma. MOLECULAR BIOSYSTEMS 2016; 12:1638-52. [PMID: 26989839 PMCID: PMC5577932 DOI: 10.1039/c5mb00637f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hemorrhagic shock, a result of extensive blood loss, is a dominant factor in battlefield morbidity and mortality. Early rodent studies in hemorrhagic shock reported carbohydrate feeding prior to the induction of hemorrhagic shock decreased mortality. When repeated in our laboratory with a porcine model, carbohydrate pre-feed resulted in a 60% increase in death rate following hemorrhagic shock with trauma when compared to fasted animals (15/32 or 47% vs. 9/32 or 28%). In an attempt to explain the unexpected death rate for pre-fed animals, we further investigated the metabolic profiles of pre-fed non-survivors (n = 15) across 4 compartments (liver, muscle, serum, and urine) at specific time intervals (pre-shock, shock, and resuscitation) and compared them to pre-fed survivors (n = 17). As hypothesized, pre-fed pigs that died as a result of hemorrhage and trauma showed differences in their metabolic and physiologic profiles at all time intervals and in all compartments when compared to pre-fed survivors. Our data suggest that, although all animals were subjected to the same shock and trauma protocol, non-survivors exhibited altered carbohydrate processing as early as the pre-shock sampling point. This was evident in (for example) the higher levels of ATP and markers of greater anabolic activity in the muscle at the pre-shock time point. Based on the metabolic findings, we propose two mechanisms that connect pre-fed status to a higher death rate: (1) animals that die are more susceptible to opening of the mitochondrial permeability transition pore, a major factor in ischemia/reperfusion injury; and (2) loss of fasting-associated survival mechanisms in pre-fed animals.
Collapse
Affiliation(s)
- Nancy E Witowski
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | | | | | - Daniel R Lexcen
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | - Kristine E Mulier
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | - Andrea Wolf
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | | | - Greg J Beilman
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
36
|
The Role of Mitochondrial Reactive Oxygen Species in Cardiovascular Injury and Protective Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8254942. [PMID: 27200148 PMCID: PMC4856919 DOI: 10.1155/2016/8254942] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ischaemia/reperfusion (I/R) injury of the heart represents a major health burden mainly associated with acute coronary syndromes. While timely coronary reperfusion has become the established routine therapy in patients with ST-elevation myocardial infarction, the restoration of blood flow into the previously ischaemic area is always accompanied by myocardial injury. The central mechanism involved in this phenomenon is represented by the excessive generation of reactive oxygen species (ROS). Besides their harmful role when highly generated during early reperfusion, minimal ROS formation during ischaemia and/or at reperfusion is critical for the redox signaling of cardioprotection. In the past decades, mitochondria have emerged as the major source of ROS as well as a critical target for cardioprotective strategies at reperfusion. Mitochondria dysfunction associated with I/R myocardial injury is further described and ultimately analyzed with respect to its role as source of both deleterious and beneficial ROS. Furthermore, the contribution of ROS in the highly investigated field of conditioning strategies is analyzed. In the end, the vascular sources of mitochondria-derived ROS are briefly reviewed.
Collapse
|
37
|
Paradis S, Charles AL, Meyer A, Lejay A, Scholey JW, Chakfé N, Zoll J, Geny B. Chronology of mitochondrial and cellular events during skeletal muscle ischemia-reperfusion. Am J Physiol Cell Physiol 2016; 310:C968-82. [PMID: 27076618 DOI: 10.1152/ajpcell.00356.2015] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Peripheral artery disease (PAD) is a common circulatory disorder of the lower limb arteries that reduces functional capacity and quality of life of patients. Despite relatively effective available treatments, PAD is a serious public health issue associated with significant morbidity and mortality. Ischemia-reperfusion (I/R) cycles during PAD are responsible for insufficient oxygen supply, mitochondriopathy, free radical production, and inflammation and lead to events that contribute to myocyte death and remote organ failure. However, the chronology of mitochondrial and cellular events during the ischemic period and at the moment of reperfusion in skeletal muscle fibers has been poorly reviewed. Thus, after a review of the basal myocyte state and normal mitochondrial biology, we discuss the physiopathology of ischemia and reperfusion at the mitochondrial and cellular levels. First we describe the chronology of the deleterious biochemical and mitochondrial mechanisms activated by I/R. Then we discuss skeletal muscle I/R injury in the muscle environment, mitochondrial dynamics, and inflammation. A better understanding of the chronology of the events underlying I/R will allow us to identify key factors in the development of this pathology and point to suitable new therapies. Emerging data on mitochondrial dynamics should help identify new molecular and therapeutic targets and develop protective strategies against PAD.
Collapse
Affiliation(s)
- Stéphanie Paradis
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France;
| | - Anne-Laure Charles
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Alain Meyer
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Anne Lejay
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; Department of Vascular Surgery and Kidney Transplantation, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; and
| | - James W Scholey
- Department of Medicine and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Nabil Chakfé
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Vascular Surgery and Kidney Transplantation, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; and
| | - Joffrey Zoll
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Bernard Geny
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| |
Collapse
|
38
|
Schulz R, Di Lisa F. Mitochondrial potassium homeostasis: a central player in cardioprotection. Cardiovasc Res 2016; 110:4-5. [DOI: 10.1093/cvr/cvw041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
39
|
Yang B, Fung A, Pac-Soo C, Ma D. Vascular surgery-related organ injury and protective strategies: update and future prospects. Br J Anaesth 2016; 117:ii32-ii43. [DOI: 10.1093/bja/aew211] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
40
|
Valls-Lacalle L, Barba I, Miró-Casas E, Alburquerque-Béjar JJ, Ruiz-Meana M, Fuertes-Agudo M, Rodríguez-Sinovas A, García-Dorado D. Succinate dehydrogenase inhibition with malonate during reperfusion reduces infarct size by preventing mitochondrial permeability transition. Cardiovasc Res 2015; 109:374-84. [DOI: 10.1093/cvr/cvv279] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/22/2015] [Indexed: 01/18/2023] Open
|
41
|
Orbegozo Cortés D, Puflea F, De Backer D, Creteur J, Vincent JL. Near infrared spectroscopy (NIRS) to assess the effects of local ischemic preconditioning in the muscle of healthy volunteers and critically ill patients. Microvasc Res 2015; 102:25-32. [PMID: 26265192 DOI: 10.1016/j.mvr.2015.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/12/2015] [Accepted: 08/06/2015] [Indexed: 12/28/2022]
Abstract
Near-infrared spectroscopy (NIRS) permits non-invasive evaluation of tissue oxygen saturation (StO2). A vascular occlusion test (VOT) produces transient controlled ischemia similar to that used in ischemic preconditioning. We hypothesized that we could evaluate local responses to ischemic preconditioning by performing repeated VOTs and observing the changes in different NIRS VOT-derived variables. In healthy volunteers (n=20), four VOTs were performed at 30-min intervals on one day and, in a second group (n=21), two VOTs with time intervals of 5, 15 or 30min were performed on 3 separate days. Two cohorts of patients, one with circulatory shock (n=23) and a hemodynamically stable group (n=20), were also studied, repeating the VOT twice with a 5-min interval. In the 1-day volunteers, there was a median decrease of 15 (6-21)% in the Desc slope (StO2 decrease during VOT) after the second VOT, but no significant change in the Asc slope (StO2 increase after VOT). In the 3-day volunteers, the Desc slope also decreased, regardless of the time interval between VOTs. There was no overall decrease in the Desc slope in either patient cohort with repeated VOTs but there was marked individual patient variability. Patients in whom the Desc slope decreased had less organ dysfunction at admission, required less norepinephrine (0.00 vs 0.08mcg/kg/min, p=0.02), less frequently had sepsis (12 vs 50%, p=0.02) and had a lower mortality (6 vs 39%, p=0.03) compared to those in whom it did not decrease. Repeated NIRS VOT can non-invasively assess the local effects of ischemic preconditioning in the muscle.
Collapse
Affiliation(s)
- Diego Orbegozo Cortés
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Florin Puflea
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Daniel De Backer
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
42
|
Sappal R, MacDougald M, Fast M, Stevens D, Kibenge F, Siah A, Kamunde C. Alterations in mitochondrial electron transport system activity in response to warm acclimation, hypoxia-reoxygenation and copper in rainbow trout, Oncorhynchus mykiss. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 165:51-63. [PMID: 26022556 DOI: 10.1016/j.aquatox.2015.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/20/2015] [Accepted: 05/16/2015] [Indexed: 06/04/2023]
Abstract
Fish expend significant amounts of energy to handle the numerous potentially stressful biotic and abiotic factors that they commonly encounter in aquatic environments. This universal requirement for energy singularizes mitochondria, the primary cellular energy transformers, as fundamental drivers of responses to environmental change. Our study probed the interacting effects of thermal stress, hypoxia-reoxygenation (HRO) and copper (Cu) exposure in rainbow trout to test the prediction that they act jointly to impair mitochondrial function. Rainbow trout were acclimated to 11 (controls) or 20°C for 2 months. Liver mitochondria were then isolated and their responses in vitro to Cu (0-20μM) without and with HRO were assessed. Sequential inhibition and activation of mitochondrial electron transport system (ETS) enzyme complexes permitted the measurement of respiratory activities supported by complex I-IV (CI-IV) in one run. The results showed that warm acclimation reduced fish and liver weights but increased mitochondrial protein indicating impairment of energy metabolism, increased synthesis of defense proteins and/or reduced liver water content. Whereas acute rise (11→20°C) in temperature increased mitochondrial oxidation rates supported by CI-IV, warm acclimation reduced the maximal (state 3) and increased the basal (state 4) respiration leading to global uncoupling of oxidative phosphorylation (OXPHOS). HRO profoundly inhibited both maximal and basal respiration rates supported by CI-IV, reduced RCR for all except CII and lowered CI:CII respiration ratio, an indication of decreased OXPHOS efficiency. The effects of Cu were less pronounced but more variable and included inhibition of CII-IV maximal respiration rates and stimulation of both CI and CIII basal respiration rates. Surprisingly, only CII and CIII indices exhibited significant 3-way interactions whereas 2-way interactions of acclimation either with Cu or HRO were portrayed mostly by CIV, and those of HRO and Cu were most common in CI and II respiratory indices. Our study suggests that warm acclimation blunts sensitivity of the ETS to temperature rise and that HRO and warm acclimation impose mitochondrial changes that sensitize the ETS to Cu. Overall, our study highlights the significance of the ETS in mitochondrial bioenergetic dysfunction caused by thermal stress, HRO and Cu exposure.
Collapse
Affiliation(s)
- Ravinder Sappal
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada; Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada
| | - Michelle MacDougald
- Faculty of Medicine, Memorial University of Newfoundland, Health Sciences Centre, Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Mark Fast
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada
| | - Fred Kibenge
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada
| | - Ahmed Siah
- British Columbia Centre for Aquatic Health Sciences, 871A Island Highway, Campbell River, BC, V9W 2C2, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada.
| |
Collapse
|
43
|
Dănilă MD, Privistirescu AI, Mirica SN, Sturza A, Ordodi V, Noveanu L, Duicu OM, Muntean DM. Acute inhibition of monoamine oxidase and ischemic preconditioning in isolated rat hearts: interference with postischemic functional recovery but no effect on infarct size reduction. Can J Physiol Pharmacol 2015; 93:819-25. [PMID: 26322912 DOI: 10.1139/cjpp-2015-0103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Monoamine oxidases (MAOs) have recently emerged as important mitochondrial sources of oxidative stress in the cardiovascular system. Generation of reactive oxygen species during the brief episodes of ischemic preconditioning (IPC) is responsible for the cardioprotection at reperfusion. The aim of this study was to assess the effects of two MAO inhibitors (clorgyline and pargyline) on the IPC-related protection in isolated rat hearts. Animals subjected to 30 min global ischemia and 120 min reperfusion were assigned to the following groups: (i) Control, no additional intervention; (ii) IPC, 3 cycles of 5 min ischemia and 5 min reperfusion before the index ischemia; (iii) IPC-clorgyline, IPC protocol bracketed for 5 min with clorgyline (50 μmol/L); (iv) IPC-pargyline, IPC protocol bracketed for 5 min with pargyline (0.5 mmol/L). The postischemic functional recovery was assessed by the left ventricular developed pressure (LVDP) and the indices of contractility (+dLVP/dt max) and relaxation (-dLVP/dt max). Infarct size (IS) was quantified by TTC staining. In both genders, IPC significantly improved functional recovery that was further enhanced in the presence of either clorgyline or pargyline. IS reduction was comparable among all the preconditioned groups, regardless of the presence of MAO inhibitors. In isolated rat hearts, acute inhibition of MAOs potentiates the IPC-induced postischemic functional recovery without interfering with the anti-necrotic protection.
Collapse
Affiliation(s)
- Maria D Dănilă
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Andreea I Privistirescu
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania
| | - Silvia N Mirica
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania
| | - Adrian Sturza
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Valentin Ordodi
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania
| | - Lavinia Noveanu
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Oana M Duicu
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Danina M Muntean
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| |
Collapse
|
44
|
Bibli SI, Andreadou I, Chatzianastasiou A, Tzimas C, Sanoudou D, Kranias E, Brouckaert P, Coletta C, Szabo C, Kremastinos DT, Iliodromitis EK, Papapetropoulos A. Cardioprotection by H2S engages a cGMP-dependent protein kinase G/phospholamban pathway. Cardiovasc Res 2015; 106:432-42. [PMID: 25870184 DOI: 10.1093/cvr/cvv129] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 04/02/2015] [Indexed: 12/29/2022] Open
Abstract
AIMS H2S is known to confer cardioprotection; however, the pathways mediating its effects in vivo remain incompletely understood. The purpose of the present study is to evaluate the contribution of cGMP-regulated pathways in the infarct-limiting effect of H2S in vivo. METHODS AND RESULTS Anaesthetized rabbits were subjected to myocardial ischaemia (I)/reperfusion (R), and infarct size was determined in control or H2S-exposed groups. The H2S donor sodium hydrosulfide (NaHS, an agent that generates H2S) increased cardiac cGMP and reduced the infarct size. The cGMP-dependent protein kinase (PKG)-I inhibitor DT2 abrogated the protective effect of NaHS, whereas the control peptide TAT or l-nitroarginine methyl ester (l-NAME) did not alter the effect of NaHS. Moreover, the KATP channel inhibitor, glibenclamide, partially reversed the effects of NaHS, whereas inhibition of mitochondrial KATP did not modify the NaHS response. NaHS enhanced phosphorylation of phospholamban (PLN), in a PKG-dependent manner. To further investigate the role of PLN in H2S-mediated cardioprotection, wild-type and PLN KO mice underwent I/R. NaHS did not exert cardioprotection in PLN KO mice. Unlike what was observed in rabbits, genetic or pharmacological inhibition of eNOS abolished the infarct-limiting effect of NaHS in mice. CONCLUSIONS Our findings demonstrate (i) that administration of NaHS induces cardioprotection via a cGMP/PKG/PLN pathway and (ii) contribution of nitric oxide to the H2S response is species-specific.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Faculty of Pharmacy, University of Athens, Panepistimiopolis, Zografou, Athens 15771, Greece
| | - Ioanna Andreadou
- Faculty of Pharmacy, University of Athens, Panepistimiopolis, Zografou, Athens 15771, Greece
| | - Athanasia Chatzianastasiou
- Faculty of Medicine, First Department of Critical Care and Pulmonary Services, Evangelismos Hospital, University of Athens, Athens, Greece
| | - Christos Tzimas
- Molecular Biology Department, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Despina Sanoudou
- Molecular Biology Department, Biomedical Research Foundation of the Academy of Athens, Athens, Greece Department of Pharmacology, Faculty of Medicine, University of Athens, Athens, Greece
| | - Evangelia Kranias
- Molecular Biology Department, Biomedical Research Foundation of the Academy of Athens, Athens, Greece Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium Department of Molecular Biomedical Research, VIB, Ghent, Belgium
| | - Ciro Coletta
- Department of Anesthesiology University of Texas Medical Branch, Galveston, TX, USA Shriners Burns Hospital for Children, Galveston, TX, USA
| | - Csaba Szabo
- Department of Anesthesiology University of Texas Medical Branch, Galveston, TX, USA Shriners Burns Hospital for Children, Galveston, TX, USA
| | - Dimitrios Th Kremastinos
- Faculty of Medicine, Second Department of Cardiology, Attikon University Hospital, University of Athens, Athens, Greece
| | - Efstathios K Iliodromitis
- Faculty of Medicine, Second Department of Cardiology, Attikon University Hospital, University of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Faculty of Pharmacy, University of Athens, Panepistimiopolis, Zografou, Athens 15771, Greece Molecular Biology Department, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
45
|
Di Lisa F, Itoh N. Cardiac Fgf21 synthesis and release: an autocrine loop for boosting up antioxidant defenses in failing hearts. Cardiovasc Res 2015; 106:1-3. [PMID: 25712960 DOI: 10.1093/cvr/cvv050] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo, 3, Padova 35131, Italy
| | - Nobuyuki Itoh
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto 606-8501, Japan
| |
Collapse
|
46
|
Ischaemic conditioning strategies reduce ischaemia/reperfusion-induced organ injury. Br J Anaesth 2015; 114:204-16. [DOI: 10.1093/bja/aeu302] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
47
|
Pagliaro P, Penna C. Redox signalling and cardioprotection: translatability and mechanism. Br J Pharmacol 2015; 172:1974-95. [PMID: 25303224 DOI: 10.1111/bph.12975] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/24/2014] [Accepted: 09/30/2014] [Indexed: 12/13/2022] Open
Abstract
The morbidity and mortality from coronary artery disease (CAD) remain significant worldwide. The treatment for acute myocardial infarction has improved over the past decades, including early reperfusion of culprit coronary arteries. Although it is mandatory to reperfuse the ischaemic territory as soon as possible, paradoxically this leads to additional myocardial injury, namely ischaemia/reperfusion (I/R) injury, in which redox stress plays a pivotal role and for which no effective therapy is currently available. In this review, we report evidence that the redox environment plays a pivotal role not only in I/R injury but also in cardioprotection. In fact, cardioprotective strategies, such as pre- and post-conditioning, result in a robust reduction in infarct size in animals and the role of redox signalling is of paramount importance in these conditioning strategies. Nitrosative signalling and cysteine redox modifications, such as S-nitrosation/S-nitrosylation, are also emerging as very important mechanisms in conditioning cardioprotection. The reasons for the switch from protective oxidative/nitrosative signalling to deleterious oxidative/nitrosative/nitrative stress are not fully understood. The complex regulation of this switch is, at least in part, responsible for the diminished or lack of cardioprotection induced by conditioning protocols observed in ageing animals and with co-morbidities as well as in humans. Therefore, it is important to understand at a mechanistic level the reasons for these differences before proposing a safe and useful transition of ischaemic or pharmacological conditioning. Indeed, more mechanistic novel therapeutic strategies are required to protect the heart from I/R injury and to improve clinical outcomes in patients with CAD.
Collapse
Affiliation(s)
- P Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, 10043, Orbassano, Turin, Italy
| | | |
Collapse
|
48
|
Balance of nitric oxide and reactive oxygen species in myocardial reperfusion injury and protection. J Cardiovasc Pharmacol 2014; 62:567-75. [PMID: 23921313 DOI: 10.1097/fjc.0b013e3182a50c45] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Depending on their concentrations, both nitric oxide (NO) and reactive oxygen species (ROS) take part either in myocardial ischemia reperfusion injury or in protection by ischemic and pharmacological preconditioning (Ipre) and postconditioning (Ipost). At the beginning of reperfusion, a transient release of NO is promptly scavenged by ROS to form the highly toxic peroxynitrite, which is responsible for a further increase of ROS through endothelial nitric oxide synthase uncoupling. The protective role of NO has suggested the use of NO donors to mimic Ipre and Ipost. However, NO donors have not always given the expected protection, possibly because they are responsible for the production of different amounts of ROS that depend on the amount of released NO. This review is focused on the role of the balance of NO and ROS in myocardial injury and its prevention by Ipre and Ipost and after the use of NO donors given with or without antioxidant compounds to mimic Ipre and Ipost.
Collapse
|
49
|
Zanetti F, Carpi A, Menabò R, Giorgio M, Schulz R, Valen G, Baysa A, Massimino ML, Sorgato MC, Bertoli A, Di Lisa F. The cellular prion protein counteracts cardiac oxidative stress. Cardiovasc Res 2014; 104:93-102. [PMID: 25139744 DOI: 10.1093/cvr/cvu194] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
AIMS The cellular prion protein, PrP(C), whose aberrant isoforms are related to prion diseases of humans and animals, has a still obscure physiological function. Having observed an increased expression of PrP(C) in two in vivo paradigms of heart remodelling, we focused on isolated mouse hearts to ascertain the capacity of PrP(C) to antagonize oxidative damage induced by ischaemic and non-ischaemic protocols. METHODS AND RESULTS Hearts isolated from mice expressing PrP(C) in variable amounts were subjected to different and complementary oxidative perfusion protocols. Accumulation of reactive oxygen species, oxidation of myofibrillar proteins, and cell death were evaluated. We found that overexpressed PrP(C) reduced oxidative stress and cell death caused by post-ischaemic reperfusion. Conversely, deletion of PrP(C) increased oxidative stress during both ischaemic preconditioning and perfusion (15 min) with H2O2. Supporting its relation with intracellular systems involved in oxidative stress, PrP(C) was found to influence the activity of catalase and, for the first time, the expression of p66(Shc), a protein implicated in oxidative stress-mediated cell death. CONCLUSIONS Our data demonstrate that PrP(C) contributes to the cardiac mechanisms antagonizing oxidative insults.
Collapse
Affiliation(s)
- Filippo Zanetti
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Andrea Carpi
- Department of Experimental Oncology, European Institute of Oncology, Milano, Italy
| | - Roberta Menabò
- CNR Institute of Neuroscience, University of Padova, Padova, Italy
| | - Marco Giorgio
- Department of Experimental Oncology, European Institute of Oncology, Milano, Italy
| | - Rainer Schulz
- Institut für Physiologie, Justus-Liebig Universität, Gießen, Germany
| | - Guro Valen
- Department of Physiology, University of Oslo, Oslo, Norway
| | - Anton Baysa
- Department of Physiology, University of Oslo, Oslo, Norway
| | | | - Maria Catia Sorgato
- Department of Biomedical Science, University of Padova, Padova, Italy CNR Institute of Neuroscience, University of Padova, Padova, Italy
| | | | - Fabio Di Lisa
- Department of Biomedical Science, University of Padova, Padova, Italy
| |
Collapse
|
50
|
Abstract
The field of mitochondrial ion channels has recently seen substantial progress, including the molecular identification of some of the channels. An integrative approach using genetics, electrophysiology, pharmacology, and cell biology to clarify the roles of these channels has thus become possible. It is by now clear that many of these channels are important for energy supply by the mitochondria and have a major impact on the fate of the entire cell as well. The purpose of this review is to provide an up-to-date overview of the electrophysiological properties, molecular identity, and pathophysiological functions of the mitochondrial ion channels studied so far and to highlight possible therapeutic perspectives based on current information.
Collapse
|