1
|
Maslov LN, Popov SV, Naryzhnaya NV, Mukhomedzyanov AV, Kurbatov BK, Derkachev IA, Boshchenko AA, Prasad NR, Ma H, Zhang Y, Sufianova GZ, Fu F, Pei JM. K ATP channels are regulators of programmed cell death and targets for the creation of novel drugs against ischemia/reperfusion cardiac injury. Fundam Clin Pharmacol 2023; 37:1020-1049. [PMID: 37218378 DOI: 10.1111/fcp.12924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/29/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND The use of percutaneous coronary intervention (PCI) in patients with ST-segment elevation myocardial infarction (STEMI) is associated with a mortality rate of 5%-7%. It is clear that there is an urgent need to develop new drugs that can effectively prevent cardiac reperfusion injury. ATP-sensitive K+ (KATP ) channel openers (KCOs) can be classified as such drugs. RESULTS KCOs prevent irreversible ischemia and reperfusion injury of the heart. KATP channel opening promotes inhibition of apoptosis, necroptosis, pyroptosis, and stimulation of autophagy. KCOs prevent the development of cardiac adverse remodeling and improve cardiac contractility in reperfusion. KCOs exhibit antiarrhythmic properties and prevent the appearance of the no-reflow phenomenon in animals with coronary artery occlusion and reperfusion. Diabetes mellitus and a cholesterol-enriched diet abolish the cardioprotective effect of KCOs. Nicorandil, a KCO, attenuates major adverse cardiovascular event and the no-reflow phenomenon, reduces infarct size, and decreases the incidence of ventricular arrhythmias in patients with acute myocardial infarction. CONCLUSION The cardioprotective effect of KCOs is mediated by the opening of mitochondrial KATP (mitoKATP ) and sarcolemmal KATP (sarcKATP ) channels, triggered free radicals' production, and kinase activation.
Collapse
Affiliation(s)
- Leonid N Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Sergey V Popov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Natalia V Naryzhnaya
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Alexandr V Mukhomedzyanov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Boris K Kurbatov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Ivan A Derkachev
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Alla A Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - N Rajendra Prasad
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, India
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Yi Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Galina Z Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen, Russia
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jian-Ming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
2
|
Chapa-Dubocq XR, Rodríguez-Graciani KM, Escobales N, Javadov S. Mitochondrial Volume Regulation and Swelling Mechanisms in Cardiomyocytes. Antioxidants (Basel) 2023; 12:1517. [PMID: 37627512 PMCID: PMC10451443 DOI: 10.3390/antiox12081517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrion, known as the "powerhouse" of the cell, regulates ion homeostasis, redox state, cell proliferation and differentiation, and lipid synthesis. The inner mitochondrial membrane (IMM) controls mitochondrial metabolism and function. It possesses high levels of proteins that account for ~70% of the membrane mass and are involved in the electron transport chain, oxidative phosphorylation, energy transfer, and ion transport, among others. The mitochondrial matrix volume plays a crucial role in IMM remodeling. Several ion transport mechanisms, particularly K+ and Ca2+, regulate matrix volume. Small increases in matrix volume through IMM alterations can activate mitochondrial respiration, whereas excessive swelling can impair the IMM topology and initiates mitochondria-mediated cell death. The opening of mitochondrial permeability transition pores, the well-characterized phenomenon with unknown molecular identity, in low- and high-conductance modes are involved in physiological and pathological increases of matrix volume. Despite extensive studies, the precise mechanisms underlying changes in matrix volume and IMM structural remodeling in response to energy and oxidative stressors remain unknown. This review summarizes and discusses previous studies on the mechanisms involved in regulating mitochondrial matrix volume, IMM remodeling, and the crosstalk between these processes.
Collapse
Affiliation(s)
| | | | | | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (N.E.)
| |
Collapse
|
3
|
Role of Oxidative Stress in Reperfusion following Myocardial Ischemia and Its Treatments. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6614009. [PMID: 34055195 PMCID: PMC8149218 DOI: 10.1155/2021/6614009] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/21/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022]
Abstract
Myocardial ischemia is a disease with high morbidity and mortality, for which reperfusion is currently the standard intervention. However, the reperfusion may lead to further myocardial damage, known as myocardial ischemia/reperfusion injury (MI/RI). Oxidative stress is one of the most important pathological mechanisms in reperfusion injury, which causes apoptosis, autophagy, inflammation, and some other damage in cardiomyocytes through multiple pathways, thus causing irreversible cardiomyocyte damage and cardiac dysfunction. This article reviews the pathological mechanisms of oxidative stress involved in reperfusion injury and the interventions for different pathways and targets, so as to form systematic treatments for oxidative stress-induced myocardial reperfusion injury and make up for the lack of monotherapy.
Collapse
|
4
|
Signaling pathways targeting mitochondrial potassium channels. Int J Biochem Cell Biol 2020; 125:105792. [PMID: 32574707 DOI: 10.1016/j.biocel.2020.105792] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
In this review, we describe key signaling pathways regulating potassium channels present in the inner mitochondrial membrane. The signaling cascades covered here include phosphorylation, redox reactions, modulation by calcium ions and nucleotides. The following types of potassium channels have been identified in the inner mitochondrial membrane of various tissues: ATP-sensitive, Ca2+-activated, voltage-gated and two-pore domain potassium channels. The direct roles of these channels involve regulation of mitochondrial respiration, membrane potential and synthesis of reactive oxygen species (ROS). Changes in channel activity lead to diverse pro-life and pro-death responses in different cell types. Hence, characterizing the signaling pathways regulating mitochondrial potassium channels will facilitate understanding the physiological role of these proteins. Additionally, we describe in this paper certain regulatory mechanisms, which are unique to mitochondrial potassium channels.
Collapse
|
5
|
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
|
6
|
Abstract
The World Health Organization suggests that the cardiovascular diseases (CVDs) are the major cause of mortality and account for two-thirds of the deaths all over the world. These diseases kill about 17 million people every year and 3 in every 10 deaths are due to these diseases. The past decade has seen considerable improvements in diagnosis as well as treatment of various heart diseases. Various new therapeutic targets are being identified through in-depth knowledge of the disease mechanisms which has favored the testing of new strategies leading to newer treatment options. Opioid peptides and G-protein-coupled opioid receptors (ORs) have been previously studied widely in terms of central nervous system actions in mitigating the pain and drug abuse. The OR agonism or antagonism induces cytoprotective states in the myocardium, rendering these receptors as an attractive target for protection of heart from the fatal heart diseases. The opioids can provide an extended window of protection of the heart from various diseases. Although the mechanisms may not be fully understood, they seem to play a crucial role in various CVDs such as hypertension, hyperlipidemia, ischemic heart disease myocardial ischemia, and congestive heart failure. Since these compounds are already being used in acute and chronic pain, soon these compounds might be approved for use as cardioprotective agents. The following review focuses on the new information acquired on the role of the ORs in various CVDs.
Collapse
Affiliation(s)
- Hemangi Rawal
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | | |
Collapse
|
7
|
Baek KI, Li R, Jen N, Choi H, Kaboodrangi A, Ping P, Liem D, Beebe T, Hsiai TK. Flow-Responsive Vascular Endothelial Growth Factor Receptor-Protein Kinase C Isoform Epsilon Signaling Mediates Glycolytic Metabolites for Vascular Repair. Antioxid Redox Signal 2018; 28:31-43. [PMID: 28762754 PMCID: PMC5695747 DOI: 10.1089/ars.2017.7044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/31/2017] [Accepted: 07/31/2017] [Indexed: 12/19/2022]
Abstract
AIMS Hemodynamic shear stress participates in maintaining vascular redox status. Elucidating flow-mediated endothelial metabolites enables us to discover metabolic biomarkers and therapeutic targets. We posited that flow-responsive vascular endothelial growth factor receptor (VEGFR)-protein kinase C isoform epsilon (PKCɛ)-6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) signaling modulates glycolytic metabolites for vascular repair. RESULTS Bidirectional oscillatory flow (oscillatory shear stress [OSS]: 0.1 ± 3 dyne·cm-2 at 1 Hz) upregulated VEGFR-dependent PKCɛ expression to a greater degree than did unidirectional pulsatile flow (pulsatile shear stress [PSS]: 23 ± 8 dyne·cm-2 at 1 Hz) in human aortic endothelial cells (p < 0.05, n = 3). PSS and OSS further upregulated PKCɛ-dependent PFKFB3 expression for glycolysis (p < 0.05, n = 4). Constitutively active PKCɛ increased, whereas dominant-negative PKCɛ reduced both basal and maximal extracellular acidification rates for glycolytic flux (p < 0.01, n = 4). Metabolomic analysis demonstrated an increase in PKCɛ-dependent glycolytic metabolite, dihydroxyacetone (DHA), but a decrease in gluconeogenic metabolite, aspartic acid (p < 0.05 vs. control, n = 6). In a New Zealand White rabbit model, both PKCɛ and PFKFB3 immunostaining was prominent in the PSS- and OSS-exposed aortic arch and descending aorta. In a transgenic Tg(flk-1:EGFP) zebrafish model, GATA-1a morpholino oligonucleotide injection (to reduce viscosity-dependent shear stress) impaired vascular regeneration after tail amputation (p < 0.01, n = 20), which was restored with PKCɛ messenger RNA (mRNA) rescue (p < 0.05, n = 5). As a corollary, siPKCɛ inhibited tube formation and vascular repair, which were restored by DHA treatment in our Matrigel and zebrafish models. Innovation and Conclusion: Flow-sensitive VEGFR-PKCɛ-PFKFB3 signaling increases the glycolytic metabolite, dihydroxyacetone, to promote vascular repair. Antioxid. Redox Signal. 28, 31-43.
Collapse
Affiliation(s)
- Kyung In Baek
- 1 Department of Bioengineering, School of Engineering and Applied Science, University of California , Los Angeles, Los Angeles, California
| | - Rongsong Li
- 2 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
| | - Nelson Jen
- 1 Department of Bioengineering, School of Engineering and Applied Science, University of California , Los Angeles, Los Angeles, California
| | - Howard Choi
- 2 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
| | - Amir Kaboodrangi
- 1 Department of Bioengineering, School of Engineering and Applied Science, University of California , Los Angeles, Los Angeles, California
| | - Peipei Ping
- 2 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
- 3 Department of Physiology, School of Medicine, University of California , Los Angeles, Los Angeles, California
| | - David Liem
- 2 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
| | - Tyler Beebe
- 1 Department of Bioengineering, School of Engineering and Applied Science, University of California , Los Angeles, Los Angeles, California
| | - Tzung K Hsiai
- 1 Department of Bioengineering, School of Engineering and Applied Science, University of California , Los Angeles, Los Angeles, California
- 2 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
- 3 Department of Physiology, School of Medicine, University of California , Los Angeles, Los Angeles, California
- 4 Greater Los Angeles VA Healthcare System , Los Angeles, California
- 5 Department of Medical Engineering, California Institute of Technology , Pasadena, California
| |
Collapse
|
8
|
Djafarzadeh S, Vuda M, Jeger V, Takala J, Jakob SM. The Effects of Fentanyl on Hepatic Mitochondrial Function. Anesth Analg 2017; 123:311-25. [PMID: 27089001 DOI: 10.1213/ane.0000000000001280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Remifentanil interferes with hepatic mitochondrial function. The aim of the present study was to evaluate whether hepatic mitochondrial function is affected by fentanyl, a more widely used opioid than remifentanil. METHODS Human hepatoma HepG2 cells were exposed to fentanyl or pretreated with naloxone (an opioid receptor antagonist) or 5-hydroxydecanoate (5-HD, an inhibitor of mitochondrial adenosine triphosphate (ATP)-sensitive potassium [mitoKATP] channels), followed by incubation with fentanyl. Mitochondrial function and metabolism were then analyzed. RESULTS Fentanyl marginally reduced maximal mitochondrial complex-specific respiration rates using exogenous substrates (decrease in medians: 11%-18%; P = 0.003-0.001) but did not affect basal cellular respiration rates (P = 0.834). The effect on stimulated respiration was prevented by preincubation with naloxone or 5-HD. Fentanyl reduced cellular ATP content in a dose-dependent manner (P < 0.001), an effect that was not significantly prevented by 5-HD and not explained by increased total ATPase concentration. However, in vitro ATPase activity of recombinant human permeability glycoprotein (an ATP-dependent drug efflux transporter) was significantly stimulated by fentanyl (P = 0.004). CONCLUSIONS Our data suggest that fentanyl reduces stimulated mitochondrial respiration of cultured human hepatocytes by a mechanism that is blocked by a mitoKATP channel antagonist. Increased energy requirements for fentanyl efflux transport may offer an explanation for the substantial decrease in cellular ATP concentration.
Collapse
Affiliation(s)
- Siamak Djafarzadeh
- From the *Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland; and †Department of Clinical Research, Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | | | | | | |
Collapse
|
9
|
Wang S, Zhang F, Zhao G, Cheng Y, Wu T, Wu B, Zhang YE. Mitochondrial PKC-ε deficiency promotes I/R-mediated myocardial injury via GSK3β-dependent mitochondrial permeability transition pore opening. J Cell Mol Med 2017; 21:2009-2021. [PMID: 28266127 PMCID: PMC5571523 DOI: 10.1111/jcmm.13121] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 01/05/2017] [Indexed: 11/29/2022] Open
Abstract
Mitochondrial fission is critically involved in cardiomyocyte apoptosis, which has been considered as one of the leading causes of ischaemia/reperfusion (I/R)‐induced myocardial injury. In our previous works, we demonstrate that aldehyde dehydrogenase‐2 (ALDH2) deficiency aggravates cardiomyocyte apoptosis and cardiac dysfunction. The aim of this study was to elucidate whether ALDH2 deficiency promotes mitochondrial injury and cardiomyocyte death in response to I/R stress and the underlying mechanism. I/R injury was induced by aortic cross‐clamping for 45 min. followed by unclamping for 24 hrs in ALDH2 knockout (ALDH2−/−) and wild‐type (WT) mice. Then myocardial infarct size, cell apoptosis and cardiac function were examined. The protein kinase C (PKC) isoform expressions and their mitochondrial translocation, the activity of dynamin‐related protein 1 (Drp1), caspase9 and caspase3 were determined by Western blot. The effects of N‐acetylcysteine (NAC) or PKC‐δ shRNA treatment on glycogen synthase kinase‐3β (GSK‐3β) activity and mitochondrial permeability transition pore (mPTP) opening were also detected. The results showed that ALDH2−/− mice exhibited increased myocardial infarct size and cardiomyocyte apoptosis, enhanced levels of cleaved caspase9, caspase3 and phosphorylated Drp1. Mitochondrial PKC‐ε translocation was lower in ALDH2−/− mice than in WT mice, and PKC‐δ was the opposite. Further data showed that mitochondrial PKC isoform ratio was regulated by cellular reactive oxygen species (ROS) level, which could be reversed by NAC pre‐treatment under I/R injury. In addition, PKC‐ε inhibition caused activation of caspase9, caspase3 and Drp1Ser616 in response to I/R stress. Importantly, expression of phosphorylated GSK‐3β (inactive form) was lower in ALDH2−/− mice than in WT mice, and both were increased by NAC pre‐treatment. I/R‐induced mitochondrial translocation of GSK‐3β was inhibited by PKC‐δ shRNA or NAC pre‐treatment. In addition, mitochondrial membrane potential (∆Ψm) was reduced in ALDH2−/− mice after I/R, which was partly reversed by the GSK‐3β inhibitor (SB216763) or PKC‐δ shRNA. Collectively, our data provide the evidence that abnormal PKC‐ε/PKC‐δ ratio promotes the activation of Drp1 signalling, caspase cascades and GSK‐3β‐dependent mPTP opening, which results in mitochondrial injury‐triggered cardiomyocyte apoptosis and myocardial dysfuction in ALDH2−/− mice following I/R stress.
Collapse
Affiliation(s)
- Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feng Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gang Zhao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Cheng
- Heart Centre of Zhengzhou Ninth People's Hospital, Zhengzhou, Henan, China
| | - Ting Wu
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Bing Wu
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - You-En Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
10
|
Capuani B, Pacifici F, Pastore D, Palmirotta R, Donadel G, Arriga R, Bellia A, Di Daniele N, Rogliani P, Abete P, Sbraccia P, Guadagni F, Lauro D, Della-Morte D. The role of epsilon PKC in acute and chronic diseases: Possible pharmacological implications of its modulators. Pharmacol Res 2016; 111:659-667. [PMID: 27461137 DOI: 10.1016/j.phrs.2016.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
Abstract
Epsilon Protein kinase C (εPCK) is a particular kinase that, when activated, is able to protect against different stress injuries and therefore has been proposed to be a potential molecular target against acute and chronic diseases. Particular attention has been focused on εPCK for its involvement in the protective mechanism of Ischemic Preconditioning (IPC), a powerful endogenous mechanism characterized by subthreshold ischemic insults able to protect organs against ischemic injury. Therefore, in the past decades several εPCK modulators have been tested with the object to emulate εPCK mediate protection. Among these the most promising, so far, has been the ΨεRACK peptide, a homologous of RACK receptor for εPKC, that when administrated can mimic its effect in the cells. However, results from studies on εPCK indicate controversial role of this kinase in different organs and diseases, such as myocardial infarct, stroke, diabetes and cancer. Therefore, in this review we provide a discussion on the function of εPCK in acute and chronic diseases and how the different activators and inhibitors have been used to modulate its activity. A better understanding of its function is still needed to definitively target εPCK as novel therapeutic strategy.
Collapse
Affiliation(s)
- Barbara Capuani
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Pacifici
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Donatella Pastore
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Giulia Donadel
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Roberto Arriga
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alfonso Bellia
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Fiorella Guadagni
- IRCCS San Raffaele Pisana, Rome, Italy; San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Davide Lauro
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - David Della-Morte
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS San Raffaele Pisana, Rome, Italy.
| |
Collapse
|
11
|
Lai LN, Zhang XJ, Zhang XY, Song LH, Guo CH, Lei JW, Song XL. Lazaroid U83836E protects the heart against ischemia reperfusion injury via inhibition of oxidative stress and activation of PKC. Mol Med Rep 2016; 13:3993-4000. [PMID: 27035121 DOI: 10.3892/mmr.2016.5030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 03/07/2016] [Indexed: 11/05/2022] Open
Abstract
Oxidative stress has been demonstrated to be important during myocardial ischemia/reperfusion injury (MIRI). The lazaroid U83836E, which combines the amino functionalities of the 21‑aminosteroids with the antioxidant ring portion of vitamin E, is a reactive oxygen species scavenger. The aim of the current study was to investigate the effect of U83836E on MIRI and its mechanisms of action. Rat hearts were subjected to 30 min ligation of the left anterior descending coronary artery, followed by 2 h reperfusion. The results demonstrated that at 5 mg/kg, U83836E markedly protected cardiac function in ischemia/reperfusion rat models, decreased the malondialdehyde content and creatinine kinase activity, while increasing superoxide dismutase and glutathione peroxidase activity. Additionally, U83836E significantly decreased the histological damage to the myocardium, reduced the area of myocardial infarction in the left ventricle and modified the mitochondrial dysfunction. Furthermore, U83836E enhanced the translocation of protein kinase Cε (PKCε) from the cytoplasm to the membrane. However, the cardioprotective effects of U83836E were reduced in the presence of the PKC inhibitor, chelerythrine (1 mg/kg). Therefore, the results of the present study suggest that U83836E has a potent protective effect against MIRI in rat models through the direct anti‑oxidative stress mechanisms and the activation of PKC signaling.
Collapse
Affiliation(s)
- Li-Na Lai
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xiao-Jing Zhang
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xiao-Yi Zhang
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Li-Hua Song
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Chun-Hua Guo
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Jing-Wen Lei
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xiao-Liang Song
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| |
Collapse
|
12
|
Abstract
KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.
Collapse
Affiliation(s)
- Monique N Foster
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - William A Coetzee
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
13
|
Kruzliak P, Pechanova O, Kara T. New perspectives of nitric oxide donors in cardiac arrest and cardiopulmonary resuscitation treatment. Heart Fail Rev 2015; 19:383-90. [PMID: 23712508 PMCID: PMC3976759 DOI: 10.1007/s10741-013-9397-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Nitric oxide (NO) is often used to treat heart failure accompanied with pulmonary edema. According to present knowledge, however, NO donors are contraindicated when systolic blood pressure is less than 90 mmHg. Based on recent findings and our own clinical experience, we formulated a hypothesis about the new breakthrough complex lifesaving effects of NO donors in patients with cardiac arrest and cardiopulmonary resuscitation therapy. It includes a direct hemodynamic effect of NO donors mediated through vasodilation of coronary arteries in cooperation with improvement of cardiac function and cardiac output through reversible inhibition of mitochondrial complex I and mitochondrial NO synthase, followed by reduction in reactive oxygen species and correction of myocardial stunning. Simultaneously, an increase in vascular sensitivity to sympathetic stimulation could lead to an increase in diastolic blood pressure. Confirmation of this hypothesis in clinical practice would mean a milestone in the treatment for cardiac arrest and cardiopulmonary resuscitation.
Collapse
Affiliation(s)
- Peter Kruzliak
- Institute of Normal and Pathological Physiology and Centre of Excellence for Regulatory Role of Nitric Oxide in Civilization Diseases, Slovak Academy of Sciences, Bratislava, Slovak Republic,
| | | | | |
Collapse
|
14
|
Malfitano C, de Souza Junior AL, Irigoyen MC. Impact of conditioning hyperglycemic on myocardial infarction rats: Cardiac cell survival factors. World J Cardiol 2014; 6:449-454. [PMID: 24976917 PMCID: PMC4072835 DOI: 10.4330/wjc.v6.i6.449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 03/14/2014] [Accepted: 04/19/2014] [Indexed: 02/06/2023] Open
Abstract
While clinical data have suggested that the diabetic heart is more susceptible to ischemic heart disease (IHD), animal data have so far pointed to a lower probability of IHD. Thus, the aim of this present review is to look at these conflicting results and discuss the protective mechanisms that conditioned hyperglycemia may confer to the heart against ischemic injury. Several mechanisms have been proposed to explain the cardioprotective action of high glucose exposure, namely, up-regulation of anti-apoptotic factor Bcl-2, inactivation of pro-apoptotic factor bad, and activation of pro-survival factors such as protein kinase B (Akt), vascular endothelial growth factor (VEGF), hypoxia inducible factor-1α and protein kinase C-ε. Indeed, cytosolic increase in Ca2+ concentration, the mitochondrial permeability transition pore, plays a key role in the genesis of ischemic injury. Previous studies have shown that the diabetic heart decreased Na+/Ca2+ and Na+/H+ exchanger activity and as such it accumulates less Ca2+ in cardiomyocyte, thus preventing cardiac injury and the associated heart dysfunctions. In addition, the expression of VEGF in diabetic animals leads to increased capillary density before myocardial infarction. Despite poor prognostic in the long-term, all these results suggest that diabetes mellitus and consequently hyperglycemia may indeed play a cardioprotective role against myocardial infarction in the short term.
Collapse
|
15
|
Lim S, Chang W, Cha MJ, Song BW, Ham O, Lee SY, Lee C, Park JH, Lee SK, Jang Y, Hwang KC. PLCδ1 protein rescues ischemia-reperfused heart by the regulation of calcium homeostasis. Mol Ther 2014; 22:1110-1121. [PMID: 24637455 PMCID: PMC4048898 DOI: 10.1038/mt.2014.46] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/09/2014] [Indexed: 01/01/2023] Open
Abstract
Myocardial Ca(2+) overload induced by ischemia/reperfusion (I/R) is a major element of myocardial dysfunction in heart failure. Phospholipase C (PLC) plays important roles in the regulation of the phosphoinositol pathway and Ca(2+) homeostasis in various types of cells. Here, we investigated the protective role of PLCδ1 against myocardial I/R injury through the regulation of Ca(2+) homeostasis. To investigate its role, PLCδ1 was fused to Hph1, a cell-permeable protein transduction domain (PTD), and treated into rat neonatal cardiomyocytes and rat hearts under respective hypoxia-reoxygenation (H/R) and ischemia-reperfusion conditions. Treatment with Hph1-PLCδ1 significantly inhibited intracellular Ca(2+) overload, reactive oxygen species generation, mitochondrial permeability transition pore opening, and mitochondrial membrane potential elevation in H/R neonatal cardiomyocytes, resulting in the inhibition of apoptosis. Intravenous injections of Hph1-PLCδ1 in rats with I/R-injured myocardium caused significant reductions in infarct size and apoptosis and also improved systolic and diastolic cardiac functioning. Furthermore, a small ions profile obtained using time-of-flight secondary ion mass spectrometry showed that treatment with Hph1-PLCδ1 leads to significant recovery of calcium-related ions toward normal levels in I/R-injured myocardium. These results suggest that Hph1-PLCδ1 may manifest as a promising cardioprotective drug due to its inhibition of the mitochondrial apoptotic pathway in cells suffering from I/R injury.
Collapse
Affiliation(s)
- Soyeon Lim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Disease, Yonsei University Health System, Seoul, Republic of Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan, Republic of Korea
| | - Min-Ji Cha
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byeong-Wook Song
- Institute of Catholic Integrative Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Incheon, Republic of Korea
| | - Onju Ham
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se-Yeon Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Changyoun Lee
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Jun-Hee Park
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Sang-Kyou Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yangsoo Jang
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ki-Chul Hwang
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Cong WT, Ling J, Tian HS, Ling R, Wang Y, Huang BB, Zhao T, Duan YM, Jin LT, Li XK. Proteomic study on the protective mechanism of fibroblast growth factor 21 to ischemia-reperfusion injury. Can J Physiol Pharmacol 2013; 91:973-84. [PMID: 24117266 DOI: 10.1139/cjpp-2012-0441] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fibroblast growth factor (FGF)-21 is a novel regulator of insulin-independent glucose transport in 3T3-L1 adipocytes and has glucose and triglyceride lowering effects in rodent models of diabetes. In this study, we found that FGF-21 can significantly attenuate ischemia-reperfusion (I/R) induced damage in H9c2 cells (rat heart). However, it is unclear which signal transduction pathway is involved in the cardioprotective effect of FGF-21. Thus, this study was designed to investigate the potential mechanism induced by FGF-21. The results showed that FGF-21 treatment prevented the oxidative stress and apoptosis associated with I/R damage by reducing the levels of superoxide anions, inhibiting glycogen synthase kinase (GSK) 3β by activating Akt phosphorylation, and recovering the levels of ATP synthase pyruvate kinase isozymes M1 and protein kinase C, thereby improving energy supply. In summary, we conclude that FGF-21 protects H9c2 cells against I/R injury mainly through the Akt-GSK-3β-caspase-3 dependent pathway, preventing oxidative stress, and recovery of the energy supply.
Collapse
Affiliation(s)
- Wei-Tao Cong
- a Zhejiang Provincial Key Laboratory of Biopharmaceuticals, Wenzhou Medical College, Wenzhou, Zhejiang 325035, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhang JY, Chen ZW, Yao H. Protective effect of urantide against ischemia-reperfusion injury via protein kinase C and phosphtidylinositol 3'-kinase - Akt pathway. Can J Physiol Pharmacol 2012; 90:637-45. [PMID: 22537485 DOI: 10.1139/y2012-048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Urantide is the most potent UT receptor antagonist compound found to date. Our previous studies have shown that it has cardioprotective effect against ischemia-reperfusion injury. However, it is unclear which signal transduction pathways are involved in the urantide-induced cardioprotective effect. This study was designed to investigate whether the effect of urantide on myocardial ischemia-reperfusion injury in rats via the protein kinase C (PKC) and phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway. The results showed that urantide at 10 and 30 µg/kg markedly inhibited the increases in serum creatine kinase fraction and lactate dehydrogenase activities and the level of cardiac troponin I, reduced the ratio of myocardial infarct size to area at risk. Urantide significantly decreased the histological damage to the myocardium and modified the ultrastructural damage in cardiac myocytes. In the presence of chelerythrine (an inhibitor of PKC, 1 mg/kg) or LY294002 (an inhibitor of PI3K-Akt, 0.3 mg/kg), the protective effect of urantide was almost completely abolished. Urantide (30 µg/kg) markedly enhanced the expression of p-Akt protein during myocardial ischemia-reperfusion injury, and this enhancement was significantly attenuated by LY294002. Therefore, our results demonstrate that urantide has a potent protective effect against myocardial ischemia-reperfusion injury in rats that may be involved with the PKC and PI3K-Akt signaling pathways.
Collapse
Affiliation(s)
- Jun-Yan Zhang
- Department of Pharmacology, Anhui Medical University, Hefei, PR China
| | | | | |
Collapse
|
18
|
Ye Z, Huang YM, Wang E, Zuo ZY, Guo QL. Sevoflurane-induced delayed neuroprotection involves mitoKATP channel opening and PKC ε activation. Mol Biol Rep 2012; 39:5049-57. [DOI: 10.1007/s11033-011-1290-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 11/30/2011] [Indexed: 11/29/2022]
|
19
|
Haider KH, Ashraf M. Preconditioning approach in stem cell therapy for the treatment of infarcted heart. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:323-56. [PMID: 22917238 DOI: 10.1016/b978-0-12-398459-3.00015-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nearly two decades of research in regenerative medicine have been focused on the development of stem cells as a therapeutic option for treatment of the ischemic heart. Given the ability of stem cells to regenerate the damaged tissue, stem-cell-based therapy is an ideal approach for cardiovascular disorders. Preclinical studies in experimental animal models and clinical trials to determine the safety and efficacy of stem cell therapy have produced encouraging results that promise angiomyogenic repair of the ischemically damaged heart. Despite these promising results, stem cell therapy is still confronted with issues ranging from uncertainty about the as-yet-undetermined "ideal" donor cell type to the nonoptimized cell delivery strategies to harness optimal clinical benefits. Moreover, these lacunae have significantly hampered the progress of the heart cell therapy approach from bench to bedside for routine clinical applications. Massive death of donor cells in the infarcted myocardium during acute phase postengraftment is one of the areas of prime concern, which immensely lowers the efficacy of the procedure. An overview of the published data relevant to stem cell therapy is provided here and the various strategies that have been adopted to develop and optimize the protocols to enhance donor stem cell survival posttransplantation are discussed, with special focus on the preconditioning approach.
Collapse
Affiliation(s)
- Khawaja Husnain Haider
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | | |
Collapse
|
20
|
Nuñez IP, Fantinelli J, Arbeláez LFG, Mosca SM. Mitochondrial KATP channels participate in the limitation of infarct size by cariporide. Naunyn Schmiedebergs Arch Pharmacol 2011; 383:563-71. [DOI: 10.1007/s00210-011-0632-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 03/27/2011] [Indexed: 12/25/2022]
|
21
|
Lemoine S, Puddu PE, Durand C, Lepage O, Babatasi G, Ivascau C, Massetti M, Gérard JL, Hanouz JL. Signaling pathways involved in postconditioning-induced cardioprotection of human myocardium, in vitro. Exp Biol Med (Maywood) 2010; 235:768-76. [PMID: 20511681 DOI: 10.1258/ebm.2010.009342] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We examined the respective role and relationship between protein kinase C (PKC), mitochondrial adenosine triphosphate-sensitive potassium (mitoK(ATP)) channel and p38 mitogen-activated protein kinase (MAPK) in postconditioning of human myocardium, in vitro. Isometrically contracting, isolated human right atrial trabeculae were exposed to 30 min hypoxia and 60 min reoxygenation. Phorbol 12-myristate 13-acetate (a PKC activator), diazoxide (a mitoK(ATP) opener) and anisomycin (a p38 MAPK activator) were superfused in early reoxygenation alone and with calphostin C (a PKC inhibitor), 5-hydroxy-decanoate (5-HD, a mitoK(ATP) channel inhibitor) and SB 202190 (a p38 MAPK inhibitor). Developed force at the end of the 60 min reoxygenation (FoC(60)) period was compared between groups (mean +/- SD). Phorbol 12-myristate 13-acetate (91 +/- 4% of baseline), diazoxide (85 +/- 5% of baseline) and anisomycin (90 +/- 4% of baseline) enhanced the FoC(60) as compared with the control group (53 +/- 7% of baseline, P < 0.0001). The enhanced FoC(60) induced by phorbol 12-myristate 13-acetate was abolished by calphostin C (52 +/- 5% of baseline) and 5-HD (56 +/- 3% of baseline), but not by SB 202190 (90 +/- 8%). The diazoxide-induced recovery of FoC(60) was attenuated by 5-HD (55 +/- 6% of baseline), but was not modified by calphostin C (87 +/- 5% of baseline) and SB 202190 (90 +/- 8% of baseline). The anisomycin-induced recovery of FoC(60) was abolished by calphostin C (61 +/- 9% of baseline) and SB 202190 (52 +/- 8% of baseline), but not by 5-HD (88 +/- 6% of baseline). In conclusion, PKC activation, opening of mitoK(ATP) channels and p38 MAPK activation in early reoxygenation induced the postconditioning of human myocardium, in vitro. Furthermore, PKC activation was upstream of the opening of mitoK(ATP) channels; p38 MAPK acted on PKC. Therefore, mitoK(ATP) and p38 MAPK seemed to be involved in two independent pathways.
Collapse
Affiliation(s)
- Sandrine Lemoine
- Laboratory of Experimental Anesthesiology and Cellular Physiology EA3212, Institut Fédératif de Recherche ICORE146 Université de Caen Basse Normandie, CHU de Caen, 14033 Caen Cedex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ng KE, Schwarzer S, Duchen MR, Tinker A. The intracellular localization and function of the ATP-sensitive K+ channel subunit Kir6.1. J Membr Biol 2010; 234:137-47. [PMID: 20306027 DOI: 10.1007/s00232-010-9241-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
Our aim was to determine the subcellular localization and functional roles of the K(ATP) channel subunit Kir6.1 in intracellular membranes. Specifically, we focused on the potential role of Kir6.1 as a subunit of the mitochondrial ATP-sensitive K+ channel. Cell imaging showed that a major proportion of heterologously expressed Kir6.1-GFP and endogenously expressed Kir6.1 was distributed in the endoplasmic reticulum with little in the mitochondria or plasma membrane. We used pharmacological and molecular tools to investigate the functional significance of this distribution. The K(ATP) channel opener diazoxide increased reactive oxygen species production, and glibenclamide abolished this effect. However, in cells lacking Kir6.1 or expressing siRNA or dominant negative constructs of Kir6.1, the same effect was seen. Ca2+ handling was examined in the muscle cell line C2C12. Transfection of the dominant negative constructs of Kir6.1 significantly reduced the amplitude and rate of rise of [Ca2+]( c ) transients elicited by ATP. This study suggests that Kir6.1 is located in the endoplasmic reticulum and plays a role in modifying Ca2+ release from intracellular stores.
Collapse
Affiliation(s)
- Keat-Eng Ng
- Deparment of Medicine, The Rayne Institute, University College London, Room 107, University Street, London, WC1E 6JF, UK
| | | | | | | |
Collapse
|
23
|
Second-generation sulfonylureas preserve inhibition of mitochondrial permeability transition by the mitochondrial K+(ATP) opener nicorandil in experimental myocardial infarction. Shock 2009; 32:247-52. [PMID: 19174741 DOI: 10.1097/shk.0b013e31819c3794] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Openers of K+(ATP) channels protect the myocardium from I/R injury. Sulfonylureas are known as potent blockers of K(ATP) channels. We investigated whether 1) mitochondrial permeability transition pore may be involved in the protection afforded by the mitoK+(ATP) opener nicorandil and 2) whether sulfonylureas may prevent this beneficial effect. Anesthetized New Zealand White rabbits underwent 30 min of coronary artery occlusion, followed by 60 (isolated mitochondria) or 240 min (infarct size) of reperfusion. They received an administration of either saline (control) or nicorandil (0.5 mg kg(-1), i.v.) 15 min before ischemia. Each control and nicorandil group was divided in four subgroups pretreated by either saline, glibenclamide (Glib; 1 mg kg(-1)), gliclazide (Glic; 1 mg kg(-1)), or glimepiride (Glim; 5 microg kg(-1)) 10 min before this. Infarct size was assessed by triphenyltetrazolium chloride staining. Mitochondria were isolated from the area at risk for further assessment of the calcium retention capacity. Glibenclamide (35 +/- 8), but neither Glic (61 +/- 9) nor Glim (48 +/- 7), reversed the improvement in calcium retention capacity due to nicorandil (58 +/- 10 vs. 27 +/- 8 nmoles CaCl2 mg(-1) proteins in control). Infarct size reduction by nicorandil (32% +/- 6% vs. 65% +/- 6% of area at risk) was abolished by Glib (55 +/- 5) but not by Glic (37 +/- 3) or Glim (31 +/- 5). These data suggest that 1) the protective effect of nicorandil involves the inhibition of the mitochondrial permeability transition pore and 2) that unlike Glib, second-generation sulfonylureas preserve this cardioprotection.
Collapse
|
24
|
Haider HK, Ashraf M. Preconditioning and stem cell survival. J Cardiovasc Transl Res 2009; 3:89-102. [PMID: 20560023 DOI: 10.1007/s12265-009-9161-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 11/24/2009] [Indexed: 01/01/2023]
Abstract
The harsh ischemic and cytokine-rich microenvironment in the infarcted myocardium, infiltrated by the inflammatory and immune cells, offers a significant challenge to the transplanted donor stem cells. Massive cell death occurs during transplantation as well as following engraftment which significantly lowers the effectiveness of the heart cell therapy. Various approaches have been adopted to overcome this problem nevertheless with multiple limitations with each of these current approaches. Cellular preconditioning and reprogramming by physical, chemical, genetic, and pharmacological manipulation of the cells has shown promise and "prime" the cells to the "state of readiness" to withstand the rigors of lethal ischemia in vitro as well as posttransplantation. This review summarizes the past and present novel approaches of ischemic preconditioning, pharmacological and genetic manipulation using preconditioning mimetics, recombinant growth factor protein treatment, and reprogramming of stem cells to overexpress survival signaling molecules, microRNAs, and trophic factors for intracrine, autocrine, and paracrine effects on cytoprotection.
Collapse
Affiliation(s)
- Husnain Kh Haider
- Department of Pathology and Laboratory Medicine, University of Cincinnati, 231-Albert, Sabin Way, OH 45267-0529, USA.
| | | |
Collapse
|
25
|
Matsumoto S, Cho S, Tosaka S, Ureshino H, Maekawa T, Hara T, Sumikawa K. Pharmacological preconditioning in type 2 diabetic rat hearts: the roles of mitochondrial ATP-sensitive potassium channels and the phosphatidylinositol 3-kinase-Akt pathway. Cardiovasc Drugs Ther 2009; 23:263-70. [PMID: 19597978 DOI: 10.1007/s10557-009-6184-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE The authors examined whether olprinone, a phosphodiesterase type 3 inhibitor, or isoflurane, a volatile anesthetic, could protect the heart against myocardial infarction in type 2 diabetic rats and whether the underlying mechanisms involve protein kinase C (PKC), mitochondrial ATP-sensitive potassium (m-K(ATP)) channels, or the phosphatidylinositol 3-kinase (PI3K)-Akt pathway. METHODS All rats underwent 30 min of coronary artery occlusion followed by 2 h of reperfusion. Wistar rats received isoflurane or olprinone before ischemia with or without the PKC inhibitor chelerythrine (CHE), the m-K(ATP) channel blocker 5-hydroxydecanoic acid (5HD), or the PI3K-Akt inhibitor LY294002 (LY). Goto-Kakizaki (GK) rats were randomly assigned to receive isoflurane or olprinone. In another group, GK rats received LY before the olprinone. RESULTS In the Wistar rats, both isoflurane (38 +/- 11%) and olprinone (40 +/- 11%) reduced infarct size as compared to the control group (59 +/- 8%). In the GK rats, olprinone (41 +/- 9%) but not isoflurane (53 +/- 11%) reduced infarct size as compared to the GK control group (58 +/- 14%). The beneficial effects of olprinone were blocked by LY (58 +/- 14%). In the Wistar rats, CHE, 5HD, and LY prevented isoflurane-induced reductions of infarct size. On the other hand, LY but not CHE or 5HD prevented olprinone-induced reductions of infarct size. CONCLUSIONS Olprinone but not isoflurane protects the heart against myocardial infarction in type 2 diabetic rats. The olprinone-induced cardioprotective effect is mediated by the PI3K-Akt pathway but not PKC or m-K(ATP) channels.
Collapse
Affiliation(s)
- Shuhei Matsumoto
- Department of Anesthesiology, Nagasaki University School of Medicine, Nagasaki, 852-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Melling CWJ, Thorp DB, Milne KJ, Noble EG. Myocardial Hsp70 phosphorylation and PKC-mediated cardioprotection following exercise. Cell Stress Chaperones 2009; 14:141-50. [PMID: 18668351 PMCID: PMC2727986 DOI: 10.1007/s12192-008-0065-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 07/04/2008] [Indexed: 10/21/2022] Open
Abstract
Both protein kinase C (PKC) activation and Hsp70 expression have been shown to be key components for exercise-mediated myocardial protection during ischemia-reperfusion injury. Given that Hsp70 has been shown to undergo inducible phosphorylation in striated muscle and liver, we hypothesized that PKC may regulate myocardial Hsp70 function and subsequent exercise-conferred cardioprotection through this phosphorylation. Hence, acute exercise of male Sprague-Dawley rats (30 m/min for 60 min at 2% grade) was employed to assess the role of PKC and its selected isoforms in phosphorylation of Hsp70 and protection of the myocardium during ischemia-reperfusion injury. It was observed that administration of the PKC inhibitor chelerythrine chloride (5 mg/kg) suppressed the activation of three exercise-induced PKC isoforms (PKCalpha, PKCdelta, and PKCepsilon) and attenuated the exercise-mediated reduction of myocardial infarct size during ischemia-reperfusion injury. While this study also demonstrated that exercise led to an alteration in the phosphorylation status of Hsp70, this posttranslational modification appeared to be dissociated from PKC activation, as exercise-induced phosphorylation of Hsp70 was unchanged following inhibition of PKC. Taken together, these results indicate that selected isoforms of PKC play an important role in exercise-mediated protection of the myocardium during ischemia-reperfusion injury. However, exercise-induced phosphorylation of Hsp70 does not appear to be a mechanism by which PKC induces this cardioprotective effect.
Collapse
Affiliation(s)
- C. W. James Melling
- School of Kinesiology, Faculty of Health Sciences, The University of Western Ontario, London, Ontario Canada N6A 3K7
| | - David B. Thorp
- School of Kinesiology, Faculty of Health Sciences, The University of Western Ontario, London, Ontario Canada N6A 3K7
| | - Kevin J. Milne
- School of Kinesiology, Faculty of Health Sciences, The University of Western Ontario, London, Ontario Canada N6A 3K7
| | - Earl G. Noble
- School of Kinesiology, Faculty of Health Sciences, The University of Western Ontario, London, Ontario Canada N6A 3K7
- Lawson Health Research Institute, The University of Western Ontario, London, Ontario Canada N6A 3K7
| |
Collapse
|
28
|
Cardioprotective effects of propofol in isolated ischemia-reperfused guinea pig hearts: role of KATP channels and GSK-3beta. Can J Anaesth 2008; 55:595-605. [PMID: 18840589 DOI: 10.1007/bf03021433] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
PURPOSE Propofol exerts cardioprotective effects, but the involved mechanisms remain obscure. The present study examines the cardioprotective effects of propofol and its role in cardiac function, including its effect on K(ATP) channel opening and the inhibition of GSK-3beta activity in ischemia-reperfused hearts. METHODS Ischemia-reperfusion (I/R) was produced in isolated guinea pig hearts by stopping coronary perfusion for 25 min, followed by reperfusion. The hearts were incubated for ten minutes, with or without propofol (25 or 50 microM), or for five minutes with 500 microM 5-hydroxydecanoate (a mitochondrial K(ATP) channel blocker) or 30 microM HMR1098 (sarcolemmal K(ATP) channel blocker), followed by five minutes with 50 microM propofol before ischemia. Action potentials on the anterior epicardial surface of the ventricle were monitored using a high-resolution charge-coupled device camera system, and at five minutes after reperfusion, GSK-3beta phosphorylation at the serine residue, Ser9, was examined. RESULTS After 35 min of reperfusion, propofol (25 and 50 microM) blunted the adverse effects of I/R and reduced infarct size (P < 0.05). In addition, prior incubation with 5-hydroxydecanoate or HMR1098 had no effect on functional recovery improved by 50 microM propofol. At five minutes after reperfusion, propofol (25 and 50 microM) shortened the duration of the action potential and increased the levels of phospho-GSK-3beta (P < 0.05). CONCLUSIONS Propofol enhanced mechanical cardiac recovery and reduced infarct size. The data further suggest that GSK-3beta play an important role in propofol cardioprotective actions during coronary reperfusion, but mitochondrial K(ATP) channels do not.
Collapse
|
29
|
Larsen JR, Aagaard S, Lie RH, Sloth E, Hasenkam JM. Sevoflurane improves myocardial ischaemic tolerance in a closed-chest porcine model. Acta Anaesthesiol Scand 2008; 52:1400-10. [PMID: 19025534 DOI: 10.1111/j.1399-6576.2008.01755.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS Volatile anaesthetics prevent experimental myocardial ischaemia-reperfusion injury (I/R) in several species, but this finding is partially inconsistent with clinical evidence. Some experimental models may not accurately represent the complex signal transduction pathways triggered by volatile anaesthetics. We therefore investigated sevoflurane I/R prevention in vivo in a porcine model with greater likeness to human physiology than models previously used and compared it with neutral anaesthetic. METHODS AND RESULTS Myocardial infarct size [IS/AAR] was compared in three groups of pigs (N=35) randomised to Control anaesthesia (pentobarbital infusion, n=12), sevoflurane inhalation alone (end-tidal concentration 3.2%) (Sevo, n=9), or both Combined (n=14), throughout ischaemia and reperfusion. Anterior/septal myocardial infarcts resulted from distal LAD coronary artery occlusion by balloon catheter for 45 min followed by 120 min of reperfusion. [IS/AAR] was measured in tetrazolium-stained heart slices after standardised image processing with computer-assisted planimetry. Measurements included full invasive monitoring. Control animals developed infarction in 55.0 +/- 3.9% (SEM) of the area at risk, Sevo in 17.5 +/- 4.4% (P=0.0002), and Combined with pentobarbital in 24.3 +/- 3.8% (P=0.0001) of the AAR, sevoflurane reducing infarct size significantly (68% and 60%, respectively). CONCLUSIONS Sevoflurane markedly decreased myocardial infarct size after prolonged coronary occlusion in a porcine model. In addition to novel sevoflurane cardioprotection in the closed-chest model, which is more comparable to normal human hearts than models previously used, sevoflurane cardioprotection is substantiated in the juvenile intact organism. The perspectives underline recommending volatile anaesthetics in risk patients and in cardiac surgery.
Collapse
Affiliation(s)
- J R Larsen
- Department of Anaesthesiology-Intensive Care, Aarhus University Hospital, Skejby, Aarhus N, Denmark.
| | | | | | | | | |
Collapse
|
30
|
Protein kinase C subtypes and retinal ischemic preconditioning. Exp Eye Res 2008; 87:300-11. [PMID: 18722601 DOI: 10.1016/j.exer.2008.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2007] [Revised: 05/19/2008] [Accepted: 05/27/2008] [Indexed: 11/23/2022]
Abstract
The purpose of our study was to determine the specific subtypes of protein kinase C involved in the neuroprotection afforded by retinal ischemic preconditioning (IPC), their relationship to the opening of mitochondrial KATP (mKATP) channels, and their role in apoptosis after preconditioning and ischemia. Rats were subjected to retinal ischemia after IPC, or retinas were rendered ischemic after pharmacological opening of mKATP channels. Using immunohistochemistry and image analysis, we determined cellular localization of PKC subtypes. We blocked PKC-delta and -epsilon to study the effect on protection with IPC or with IPC-mimicking by the opening of mKATP channels. PKC subtypes were inhibited pharmacologically or with interfering RNA. Electroretinography assessed functional recovery after ischemia. IPC was effectively mimicked by injection of diazoxide to open the mKATP channel. IPC and/or its mimicking were attenuated by the PKC-delta inhibitor rottlerin and by interfering RNA targeting PKC-delta or -epsilon. Using TUNEL staining and Western blotting for caspase-3 and fodrin breakdown we assessed apoptosis. The injection of interfering RNA to PKC-delta and -epsilon before preconditioning significantly enhanced TUNEL staining as well as the cleavage of caspase-3 and fodrin after ischemia. In summary, our experiments have shown that both PKC-delta and -epsilon subtypes are involved in the cellular signaling that results in neuroprotection from IPC and that both are downstream of the opening of mKATP channels.
Collapse
|
31
|
Clarke SJ, Khaliulin I, Das M, Parker JE, Heesom KJ, Halestrap AP. Inhibition of mitochondrial permeability transition pore opening by ischemic preconditioning is probably mediated by reduction of oxidative stress rather than mitochondrial protein phosphorylation. Circ Res 2008; 102:1082-90. [PMID: 18356542 DOI: 10.1161/circresaha.107.167072] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inhibition of mitochondrial permeability transition pore (MPTP) opening at reperfusion is critical for cardioprotection by ischemic preconditioning (IP). Some studies have implicated mitochondrial protein phosphorylation in this effect. Here we confirm that mitochondria rapidly isolated from preischemic control and IP hearts show no significant difference in calcium-mediated MPTP opening, whereas IP inhibits MPTP opening in mitochondria isolated from IP hearts following 30 minutes of global normothermic ischemia or 3 minutes of reperfusion. Analysis of protein phosphorylation in density-gradient purified mitochondria was performed using both 2D and 1D electrophoresis, with detection of phosphoproteins using Pro-Q Diamond or phospho-amino-specific antibodies. Several phosphoproteins were detected, including voltage-dependent anion channels isoforms 1 and 2, but none showed significant IP-mediated changes either before ischemia or during ischemia and reperfusion, and neither Western blotting nor 2D fluorescence difference gel electrophoresis detected translocation of protein kinase C (alpha, epsilon, or delta isoforms), glycogen synthase kinase 3beta, or Akt to the mitochondria following IP. In freeze-clamped hearts, changes in phosphorylation of GSK3beta, Akt, and AMP-activated protein kinase were detected following ischemia and reperfusion but no IP-mediated changes correlated with MPTP inhibition or cardioprotection. However, measurement of mitochondrial protein carbonylation, a surrogate marker for oxidative stress, suggested that a reduction in mitochondrial oxidative stress at the end of ischemia and during reperfusion may account for IP-mediated inhibition of MPTP. The signaling pathways mediating this effect and maintaining it during reperfusion are discussed.
Collapse
Affiliation(s)
- Samantha J Clarke
- Department of Biochemistry and the Bristol Heart Institute, University of Bristol, Bristol BS8 1TD, United Kingdom
| | | | | | | | | | | |
Collapse
|
32
|
Bouwman RA, Musters RJP, van Beek-Harmsen BJ, de Lange JJ, Lamberts RR, Loer SA, Boer C. Sevoflurane-induced cardioprotection depends on PKC-alpha activation via production of reactive oxygen species. Br J Anaesth 2007; 99:639-45. [PMID: 17905752 DOI: 10.1093/bja/aem202] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND We previously demonstrated the involvement of the Ca2+-independent protein kinase C-delta (PKC-delta) isoform in sevoflurane-induced cardioprotection against ischaemia and reperfusion (I/R) injury. Since sevoflurane is known to modulate myocardial Ca2+-handling directly, in this study we investigated the role of the Ca2+-dependent PKC-alpha isoform in sevoflurane-induced cardioprotective signalling in relation to reactive oxygen species (ROS), adenosine triphosphate-sensitive mitochondrial K+ (mitoK+(ATP)) channels, and PKC-delta. METHODS Preconditioned (15 min 3.8 vol% sevoflurane) isolated rat right ventricular trabeculae were subjected to I/R, consisting of 40 min superfusion with hypoxic, glucose-free buffer, followed by normoxic glucose-containing buffer for 60 min. After reperfusion, contractile recovery was expressed as percentage of force development before I/R. The role of PKC-alpha, ROS, mitoK+(ATP) channels, and PKC-delta was established using the following pharmacological inhibitors: Go6976 (GO; 50 nM), n-(2-mercaptopropionyl)-glycine (MPG; 300 microM), 5-hydroxydecanoic acid sodium (5HD; 100 microM), and rottlerin (ROT; 1 microM). RESULTS Preconditioning of trabeculae with sevoflurane improved contractile recovery after I/R [65 (3)% (I/R + SEVO) vs 47 (3)% (I/R); n = 8; P < 0.05]. This cardioprotective effect was attenuated in trabeculae treated with GO [42 (4)% (I/R + SEVO + GO); P > 0.05 vs (I/R)]. In sevoflurane-treated trabeculae, PKC-alpha translocated towards mitochondria, as shown by immunofluorescent co-localization analysis. GO and MPG, but not 5HD or ROT, abolished this translocation. CONCLUSIONS Sevoflurane improves post-ischaemic contractile recovery via activation of PKC-alpha. ROS production, but not opening of mitoK+(ATP) channels, precedes PKC-alpha translocation towards mitochondria. This study shows the involvement of Ca2+-dependent PKC-alpha in addition to the well-established role of Ca2+-independent PKC isoforms in sevoflurane-induced cardioprotection.
Collapse
Affiliation(s)
- R A Bouwman
- VU University Medical Center (VUMC), Department of Anaesthesiology, Institute for Cardiovascular Research Vrije Universiteit (ICaR-VU), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
33
|
Takahashi Y, Takemura S, Minamiyama Y, Shibata T, Hirai H, Sasaki Y, Sakaguchi M, Suehiro S. Landiolol has cardioprotective effects against reperfusion injury in the rat heart via the PKCepsilon signaling pathway. Free Radic Res 2007; 41:757-69. [PMID: 17577736 DOI: 10.1080/10715760701338810] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Landiolol, a highly cardioselective beta1-blocker, has cardioprotective effects against ischemia-reperfusion injury, although the precise mechanism is still unclear. The aim of this study was to clarify the cardioprotective mechanism of landiolol. Experiments were performed on Langendorff-perfused rat hearts undergoing 20 min stabilization, and 45 min of ischemia followed by 60 min of reperfusion. Various drugs with or without landiolol (100 microM) were administered before ischemia for 20 min. Preischemic administration of landiolol reduced cardiac cellular damage and improved the recovery of cardiac function by about 40%. The alpha1 blocker prazosin, the protein kinase C (PKC) inhibitor chelerythrine or the K(ATP) channel blocker glibenclamide, but not the selective mitochondrial K(ATP) channel blocker 5-hydroxydecanoate abrogated the cardioprotective effect induced by landiolol. Following landiolol pretreatment the activation of PKCepsilon and heat shock protein 27 were significantly higher than that in control. These data indicate that preischemic application of landiolol induces cardioprotective effects through PKCepsilon-mediated pathway, similar to that afforded by ischemic preconditioning.
Collapse
Affiliation(s)
- Yosuke Takahashi
- Department of Cardiovascular Surgery, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Gao J, Chua CC, Chen Z, Wang H, Xu X, Hamdy RC, McMullen JR, Shioi T, Izumo S, Chua BH. Resistin, an adipocytokine, offers protection against acute myocardial infarction. J Mol Cell Cardiol 2007; 43:601-9. [PMID: 17904155 PMCID: PMC2692307 DOI: 10.1016/j.yjmcc.2007.08.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 08/09/2007] [Indexed: 11/15/2022]
Abstract
Resistin, an adipocyte-derived hormone, is thought to represent a link between obesity and insulin-resistant diabetes. The potential role of resistin as a cardioprotective agent has not been explored. Our hypothesis is that resistin has a cardioprotective effect that is mediated by the resistin receptor-coupled activation of PI3K/Akt/PKC/K(ATP) dependent pathways. Our studies demonstrated that pretreatment of mouse hearts with 10 nM resistin for 5 min protected the heart against I/R injury in a mouse heart perfusion model. When mouse hearts were subjected to 60 min of LAD ligation followed by 4 h of reperfusion, resistin pretreatment (33 microg/kg) for 30 min or 24 h before ligation was able to significantly reduce the infarct size/risk area. The protective effect of resistin was abolished by wortmannin, as well as by an Akt inhibitor, triciribine. Resistin's protective effect was absent in Akt kinase-deficient mutant mice. The protective effect was also blocked by chelerythrine, a PKC inhibitor, and epsilonV1-2, a PKCepsilon inhibitor. Finally, the protective effect was blocked by 5-hydroxydecanoate, which blocks the opening of mitoK(ATP) channels. Resistin-induced Akt phosphorylation in HL-1 cells was inhibited by wortmannin and triciribine. Resistin also induced PKCepsilon phosphorylation, which was blocked by triciribine. These studies demonstrate that resistin's cardioprotective effect is mediated by PI3K/Akt/PKC dependent pathways. In addition to cardiomyocytes, resistin also induced Akt phosphorylation in endothelial cells and smooth muscle cells, suggesting that resistin receptors are present in these cells. The effect of resistin on apoptosis was assessed in hearts subjected to 30 min of ischemia and 3 h of reperfusion. There were significantly fewer in situ oligo ligation-positive myocyte nuclei in mice treated with resistin. Our results show that resistin can dramatically reduce apoptosis and infarct size, thus protecting the heart against I/R injury.
Collapse
Affiliation(s)
- Jinping Gao
- Cecile Cox Quillen Laboratory of Geriatrics, James H. Quillen College of Medicine, East Tennessee State University, and James H. Quillen Veterans Affairs Medical Center, Johnson City, TN 37614
| | - Chu Chang Chua
- Cecile Cox Quillen Laboratory of Geriatrics, James H. Quillen College of Medicine, East Tennessee State University, and James H. Quillen Veterans Affairs Medical Center, Johnson City, TN 37614
| | - Zhongyi Chen
- Cecile Cox Quillen Laboratory of Geriatrics, James H. Quillen College of Medicine, East Tennessee State University, and James H. Quillen Veterans Affairs Medical Center, Johnson City, TN 37614
| | - Hong Wang
- Cecile Cox Quillen Laboratory of Geriatrics, James H. Quillen College of Medicine, East Tennessee State University, and James H. Quillen Veterans Affairs Medical Center, Johnson City, TN 37614
| | - Xingshun Xu
- Cecile Cox Quillen Laboratory of Geriatrics, James H. Quillen College of Medicine, East Tennessee State University, and James H. Quillen Veterans Affairs Medical Center, Johnson City, TN 37614
| | - Ronald C. Hamdy
- Cecile Cox Quillen Laboratory of Geriatrics, James H. Quillen College of Medicine, East Tennessee State University, and James H. Quillen Veterans Affairs Medical Center, Johnson City, TN 37614
| | - Julie R. McMullen
- Cardiovascular Division, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Tetsuo Shioi
- Cardiovascular Division, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Seigo Izumo
- Cardiovascular Division, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Balvin H.L. Chua
- Cecile Cox Quillen Laboratory of Geriatrics, James H. Quillen College of Medicine, East Tennessee State University, and James H. Quillen Veterans Affairs Medical Center, Johnson City, TN 37614
| |
Collapse
|
35
|
Abstract
The purpose of this review is to describe in more detail ischemia reperfusion injury and preconditioning, and to speculate on the potential role of preconditioning in the care of critically ill patients. Current hemodynamic treatment of hypotension and hypoperfusion in critically ill patients is directed at ensuring essential organ perfusion by maintaining intravascular volume and cardiac output, and ensuring adequate oxygen delivery by maintaining arterial oxygen partial pressure and hemoglobin levels. However, morbidity and mortality remain high and new approaches to critically ill patients are required. Treatments are needed that can protect against organ ischemia during periods of low blood flow. In recent years, there has been a growing appreciation of the importance of ischemia reperfusion injury. Ischemia associated with reperfusion may result in greater injury than ischemia alone. Ischemic preconditioning is used to describe the protective effect of short periods of ischemia to an organ or tissue against longer periods of ischemia. Although first described in the myocardium, there is now evidence that this phenomenon occurs in a wide variety of organs and tissues, including the brain and other nervous tissue such as the retina and spinal cord, liver, stomach, intestines, kidney, and the lungs. Preconditioning therapy may offer a new avenue of treatment in critically ill patients. Both traditional preconditioning methods and pharmacologic agents that mimic or induce such preconditioning may be used in the future. Clinical trials of pharmacologic agents are underway in patients with coronary artery disease. Further trials of such methods and agents are needed in critically ill patients suffering from sepsis or multiorgan system failure.
Collapse
Affiliation(s)
- Peter Rock
- Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | |
Collapse
|
36
|
Garg V, Hu K. Protein kinase C isoform-dependent modulation of ATP-sensitive K+ channels in mitochondrial inner membrane. Am J Physiol Heart Circ Physiol 2007; 293:H322-32. [PMID: 17351068 DOI: 10.1152/ajpheart.01035.2006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ATP-sensitive K(+) (K(ATP)) channels in both sarcolemmal (sarcK(ATP)) and mitochondrial inner membrane (mitoK(ATP)) are the critical mediators in cellular protection of ischemic preconditioning (IPC). Whereas cardiac sarcK(ATP) contains Kir6.2 and sulfonylurea receptor (SUR)2A, the molecular identity of mitoK(ATP) remains elusive. In the present study, we tested the hypothesis that protein kinase C (PKC) may promote import of Kir6.2-containing K(ATP) into mitochondria. Fluorescence imaging of isolated mitochondria from both rat adult cardiomyocytes and COS-7 cells expressing recombinant Kir6.2/SUR2A showed that Kir6.2-containing K(ATP) channels were localized in mitochondria and this mitochondrial localization was significantly increased by PKC activation with phorbol 12-myristate 13-acetate (PMA). Fluorescence resonance energy transfer microscopy further revealed that a significant number of Kir6.2-containing K(ATP) channels were localized in mitochondrial inner membrane after PKC activation. These results were supported by Western blotting showing that the Kir6.2 protein level in mitochondria from COS-7 cells transfected with Kir6.2/SUR2A was enhanced after PMA treatment and this increase was inhibited by the selective PKC inhibitor chelerythrine. Furthermore, functional analysis indicated that the number of functional K(ATP) channels in mitochondria was significantly increased by PMA, as shown by K(ATP)-dependent decrease in mitochondrial membrane potential in COS-7 cells transfected with Kir6.2/SUR2A but not empty vector. Importantly, PKC-mediated increase in mitochondrial Kir6.2-containing K(ATP) channels was blocked by a selective PKCepsilon inhibitor peptide in both COS-7 cells and cardiomyocytes. We conclude that the K(ATP) channel pore-forming subunit Kir6.2 is indeed localized in mitochondria and that the Kir6.2 content in mitochondria is increased by activation of PKCepsilon. PKC isoform-regulated mitochondrial import of K(ATP) channels may have significant implication in cardioprotection of IPC.
Collapse
Affiliation(s)
- Vivek Garg
- Division of Pharmacology, College of Pharmacy, Ohio State University, 500 W. 12th Avenue, Columbus, OH 43210, USA
| | | |
Collapse
|
37
|
Wang Y, Ahmad N, Wang B, Ashraf M. Chronic preconditioning: a novel approach for cardiac protection. Am J Physiol Heart Circ Physiol 2007; 292:H2300-5. [PMID: 17208991 DOI: 10.1152/ajpheart.01163.2006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ischemic preconditioning is the most powerful protective mechanism known against lethal ischemia. Unfortunately, the protection lasts for only a few hours. Here we tested the hypothesis that the heart can be kept in a preconditioned state for constant protection against ischemia. In this study we chose BMS-191095 (BMS), a highly selective opener of mitochondrial ATP-sensitive K(+) (mitoK(ATP)) channels. BMS (1 mg/kg ip) was administered to rats every 24 h until 96 h. In other groups, BMS plus wortmannin (WTN, 15 microg/kg ip), an inhibitor of the phosphatidylinositol 3-kinase (PI3-K), or BMS plus 5-hydroxydecanoic acid (5-HD, 5 mg/kg ip), an inhibitor of mitoK(ATP), or BMS plus N(omega)-nitro-L-arginine methyl ester (L-NAME) (30 microg/kg ip), an inhibitor of nitric oxide (NO) synthase, were administered to rats. Rats were then subjected to 30-min left anterior descending coronary artery occlusion and 120-min reperfusion. Cardiac function, infarct size, pathological changes, and apoptosis were assessed at the end of treatments. Saline-treated hearts displayed marked contractile dysfunction and underwent pathological changes. BMS-treated rats showed significant improvement in cardiac function, and infarct size was significantly reduced in BMS-treated hearts. However, protection by BMS was abolished by 5-HD, WTN, or L-NAME. These data demonstrate that hearts can be chronically preconditioned and retain their ability to remain resistant against lethal ischemia and that this protection is mediated by activation of mitoK(ATP) via NO and PI3-K/Akt signaling pathways.
Collapse
Affiliation(s)
- Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267-0529, USA
| | | | | | | |
Collapse
|
38
|
Kim MY, Kim MJ, Yoon IS, Ahn JH, Lee SH, Baik EJ, Moon CH, Jung YS. Diazoxide acts more as a PKC-epsilon activator, and indirectly activates the mitochondrial K(ATP) channel conferring cardioprotection against hypoxic injury. Br J Pharmacol 2006; 149:1059-70. [PMID: 17043673 PMCID: PMC2014640 DOI: 10.1038/sj.bjp.0706922] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND AND PURPOSE Diazoxide, a well-known opener of the mitochondrial ATP-sensitive potassium (mitoK(ATP)) channel, has been demonstrated to exert cardioprotective effect against ischemic injury through the mitoK(ATP) channel and protein kinase C (PKC). We aimed to clarify the role of PKC isoforms and the relationship between the PKC isoforms and the mitoK(ATP) channel in diazoxide-induced cardioprotection. EXPERIMENTAL APPROACH In H9c2 cells and neonatal rat cardiomyocytes, PKC-epsilon activation was examined by Western blotting and kinase assay. Flavoprotein fluorescence, mitochondrial Ca(2+) and mitochondrial membrane potential were measured by confocal microscopy. Cell death was determined by TUNEL assay. KEY RESULTS Diazoxide (100 microM) induced translocation of PKC-epsilon from the cytosolic to the mitochondrial fraction. Specific blockade of PKC-epsilon by either epsilonV1-2 or dominant negative mutant PKC-epsilon (PKC-epsilon KR) abolished the anti-apoptotic effect of diazoxide. Diazoxide-induced flavoprotein oxidation was inhibited by either epsilonV1-2 or PKC-epsilon KR transfection. Treatment with 5-hydroxydecanoate (5-HD) did not affect translocation and activation of PKC-epsilon induced by diazoxide. Transfection with wild type PKC-epsilon mimicked the flavoprotein-oxidizing effect of diazoxide, and this effect was completely blocked by epsilonV1-2 or 5-HD. Diazoxide prevented the increase in mitochondrial Ca(2+), mitochondrial depolarization and cytochrome c release induced by hypoxia and all these effects of diazoxide were blocked by epsilonV1-2 or 5-HD. CONCLUSIONS AND IMPLICATIONS Diazoxide induced isoform-specific translocation of PKC-epsilon as an upstream signaling molecule for the mitoK(ATP) channel, rendering cardiomyocytes resistant to hypoxic injury through inhibition of the mitochondrial death pathway.
Collapse
Affiliation(s)
- M-Y Kim
- Department of Physiology, School of Medicine, Ajou University, Suwon, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Ren B, Shen Y, Shao H, Qian J, Wu H, Jing H. Brain natriuretic peptide limits myocardial infarct size dependent of nitric oxide synthase in rats. Clin Chim Acta 2006; 377:83-7. [PMID: 17026975 DOI: 10.1016/j.cca.2006.08.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2006] [Revised: 08/21/2006] [Accepted: 08/24/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND Brain natriuretic peptide (BNP) has recently been shown to have a cardioprotective effect in animal models of myocardial ischemia-reperfusion (I-R) injury. We hypothesized that exogenous BNP limits myocardial infarction on nitric oxide synthase pathway. METHODS A rat model of myocardial I-R injury was established by ligating the left descending coronary artery for 30 min and then reperfusing for 2 h. BNP was injected with different dose 5 min after the ligation and lasting for 145 min. The myocardial infarct size and the area at risk of ischemia were measured by staining with triphenyltetrazolium chloride (TTC) and Evans blue dye. To examine the role of nitric oxide synthase (NOS), expression of eNOS in the left ventricle was analyzed by western blotting. Nomega-nitro-L-arginine methyl ester (L-NAME; 30 ug/kg), or S-methylisothiourea (SMT; 3 ug/kg) was administrated before I-R with or without BNP. RESULTS The control infarct-to-risk ratio was 45.1+/-1.72% (means+/-SE). BNP infused 5 min after ischemia limited infarct size in a dosage-dependent manner, with maximal protection observed at 0.01 ug/(kg min) (infarct-to-risk: 24.7+/-1.69%, P<0.01 vs. control), associated with a 10-fold increase of myocardial endothelial nitric oxide synthase above the control value. Protection afforded by BNP was abolished by L-NAME but not by SMT, suggesting the involvement of putative endothelial but not inducible nitric oxide synthase activation. CONCLUSIONS We conclude that natriuretic peptide/NOS/NO signaling may constitute an important injury-limiting mechanism in myocardium.
Collapse
Affiliation(s)
- Binhui Ren
- Department of Thoracic and Cardiovascular Surgery, Jinling Hospital, Clinical Medicine School of Nanjing University, Nanjing 210002, China.
| | | | | | | | | | | |
Collapse
|
40
|
Park YS, Hur EM, Choi BH, Kwak E, Jun DJ, Park SJ, Kim KT. Involvement of protein kinase C-epsilon in activity-dependent potentiation of large dense-core vesicle exocytosis in chromaffin cells. J Neurosci 2006; 26:8999-9005. [PMID: 16943556 PMCID: PMC6675348 DOI: 10.1523/jneurosci.2828-06.2006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neurotransmitter release is modulated in an activity-dependent manner. We showed previously that repetitive stimulation of nicotinic acetylcholine receptor (nAChR) induced activity-dependent potentiation (ADP) of large dense-core vesicle (LDCV) exocytosis in chromaffin cells. Here we report that protein kinase C (PKC)-epsilon is critically involved in ADP. Stimulation of nAChR induced activation of PKC-epsilon, and inhibition of PKC-epsilon by expression of the dominant-negative mutant of PKC-epsilon (DN-PKC-epsilon) or short interfering (siRNA) against PKC-epsilon abolished ADP via decreasing the frequency and quantal size of fused vesicles without affecting basal exocytosis, suggesting that PKC-epsilon is specifically involved in ADP. Electron microscopy revealed that inhibition of PKC-epsilon disrupts activity-induced vesicle translocation required for ADP. We also suggest the involvement of myristoylated alanine-rich C kinase substrate (MARCKS), which is known as a downstream target of PKC-epsilon, in ADP of LDCV exocytosis. The level of phospho-MARCKS correlated with the time course of ADP and was reduced by transfection with DN-PKC-epsilon. Actin filament disassembly induced by MARCKS phosphorylation was also significantly blocked by transfection of DN-PKC-epsilon. Furthermore, knockdown of MARCKS by siRNA resulted in inhibition of ADP and reduction of the number of fused vesicles. Together, we provide evidence that ADP of LDCV exocytosis is regulated by PKC-epsilon and its downstream target MARCKS via modulating vesicle translocation.
Collapse
Affiliation(s)
- Yong-Soo Park
- Department of Life Science, Division of Molecular and Life Sciences, Systems Biodynamics National Core Research Center, Pohang University of Science and Technology, Pohang, 790-784, South Korea, and
| | - Eun-Mi Hur
- Department of Life Science, Division of Molecular and Life Sciences, Systems Biodynamics National Core Research Center, Pohang University of Science and Technology, Pohang, 790-784, South Korea, and
| | - Bo-Hwa Choi
- Department of Life Science, Division of Molecular and Life Sciences, Systems Biodynamics National Core Research Center, Pohang University of Science and Technology, Pohang, 790-784, South Korea, and
| | - Eunyee Kwak
- Department of Life Science, Division of Molecular and Life Sciences, Systems Biodynamics National Core Research Center, Pohang University of Science and Technology, Pohang, 790-784, South Korea, and
| | - Dong-Jae Jun
- Department of Life Science, Division of Molecular and Life Sciences, Systems Biodynamics National Core Research Center, Pohang University of Science and Technology, Pohang, 790-784, South Korea, and
| | - Su-Jin Park
- Microscopy and Imaging System, Carl Zeiss Company, Seoul, 121-828, South Korea
| | - Kyong-Tai Kim
- Department of Life Science, Division of Molecular and Life Sciences, Systems Biodynamics National Core Research Center, Pohang University of Science and Technology, Pohang, 790-784, South Korea, and
| |
Collapse
|
41
|
Cao CM, Yan WY, Liu J, Kam KWL, Zhan SZ, Sham JSK, Wong TM. Attenuation of mitochondrial, but not cytosolic, Ca2+ overload reduces myocardial injury induced by ischemia and reperfusion. Acta Pharmacol Sin 2006; 27:911-8. [PMID: 16787576 DOI: 10.1111/j.1745-7254.2006.00391.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM Attenuation of mitochondrial Ca2+ ([Ca2+]m), but not cytosolic Ca2+ ([Ca2+]c), overload improves contractile recovery. We hypothesized that attenuation of [Ca2+]m, but not [Ca2+]c, overload confers cardioprotection against ischemia/reperfusion-induced injury. METHODS Infarct size from isolated perfused rat heart, cell viability, and electrically-induced Ca2+ transient in isolated rat ventricular myocytes were measured. We determined the effects of BAPTA-AM, a Ca2+ chelator, at concentrations that abolish the overload of both [Ca2+]c and [Ca2+]m, and ruthenium red, an inhibitor of mitochondrial uniporter of Ca2+ transport, at concentrations that abolish the overload of [Ca2+]m, but not [Ca2+]c, on cardiac injury induced by ischemia/reperfusion. RESULTS Attenuation of both [Ca2+]m and [Ca2+]c by BAPTA-AM, and attenuation of [Ca2+]m, but not [Ca2+]c, overload by ruthenium red, reduced the cardiac injury observations, indicating the importance of [Ca2+]m in cardioprotection and contractile recovery in response to ischemia/reperfusion. CONCLUSION The study has provided unequivocal evidence using a cause-effect approach that attenuation of [Ca2+]m, but not [Ca2+]c, overload is responsible for cardioprotection against ischemia/reperfusion-induced injury. We also confirmed the previous observation that attenuation of [Ca2+]m, but not [Ca2+]c, by ruthenium red improves contractile recovery following ischemia/reperfusion.
Collapse
Affiliation(s)
- Chun-mei Cao
- The Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | | | | | | | | | | | | |
Collapse
|
42
|
Yogaratnam JZ, Laden G, Madden LA, Seymour AM, Guvendik L, Cowen M, Greenman J, Cale A, Griffin S. Hyperbaric oxygen: a new drug in myocardial revascularization and protection? CARDIOVASCULAR REVASCULARIZATION MEDICINE 2006; 7:146-54. [PMID: 16945821 DOI: 10.1016/j.carrev.2006.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2006] [Accepted: 04/17/2006] [Indexed: 11/19/2022]
Abstract
Ischemia-reperfusion injury (IRI) occurs following coronary artery revascularization. Reactive oxygen species (ROS) were initially thought to play a role in the pathogenesis of this injury. However, the evidence for this is inconclusive. Recent studies involving ischemic preconditioning have identified ROS as potential mediators for the cardioprotective effects observed following this technique. Furthermore, cardiac studies involving IRI and the use of hyperbaric oxygen (HBO) have demonstrated the ability of HBO to induce cardioprotection and to attenuate IRI. This review suggests the possible role for HBO as a new drug in the arena of myocardial revascularization and cellular protection. While there is mounting clinical evidence for this, a methodological understanding of HBO's cellular mechanisms of actions appears to be lacking. As such, this article attempts to draw the similarity between HBO and other protective oxidative stress mechanisms and then to speculate in an evidence-based manner its possible cellular mechanistic role as a drug via the generation of ROS.
Collapse
Affiliation(s)
- Jeysen Zivan Yogaratnam
- Department of Cardiothoracic Surgery, Castle Hill Hospital, Castle Road, Cottingham HU16 5JQ, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Grover GJ. Mitochondrial ATP-sensitive potassium channels and mitochondrial protein kinase C: sometimes it's good to have a close neighbor. Am J Physiol Heart Circ Physiol 2006; 290:H1752-3. [PMID: 16603704 DOI: 10.1152/ajpheart.00015.2006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
44
|
Vohra HA, Fowler AG, Galiñanes M. Preconditioning with Cardioplegia Is More Effective in Reducing Apoptosis Than Is Preconditioning with Ischemia in the Human Myocardium. Ann N Y Acad Sci 2006; 1010:721-7. [PMID: 15033817 DOI: 10.1196/annals.1299.130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Hunaid A Vohra
- Department of Integrative Human Cardiovascular Physiology and Cardiac Surgery, Glenfield Hospital, Leicester LE3 9QP, UK
| | | | | |
Collapse
|
45
|
Jiang MT, Ljubkovic M, Nakae Y, Shi Y, Kwok WM, Stowe DF, Bosnjak ZJ. Characterization of human cardiac mitochondrial ATP-sensitive potassium channel and its regulation by phorbol ester in vitro. Am J Physiol Heart Circ Physiol 2005; 290:H1770-6. [PMID: 16361367 DOI: 10.1152/ajpheart.01084.2005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of the mitochondrial ATP-sensitive K+ channel (mitoKATP) and its regulation by PKC are critical events in preconditioning induced by ischemia or pharmaceutical agents in animals and humans. The properties of the human cardiac mitoKATP channel are unknown. Furthermore, there is no evidence that cytosolic PKC can directly regulate the mitoKATP channel located in the inner mitochondrial membrane (IMM) due to the physical barrier of the outer mitochondrial membrane. In the present study, we characterized the human cardiac mitoKATP channel and its potential regulation by PKC associated with the IMM. IMM fractions isolated from human left ventricles were fused into lipid bilayers in symmetrical potassium glutamate (150 mM). The conductance of native mitoKATP channels was usually below 80 pS ( approximately 70%), which was reduced by ATP and 5-hydroxydecanoic acid (5-HD) in a dose- and time-dependent manner. The native mitoKATP channel is activated by diazoxide and inhibited by ATP and 5-HD. The PKC activator phorbol 12-myristate 13-acetate (2 microM) increased the cumulative open probability of the mitoKATP channel previously inhibited by ATP (P < 0.05), but its inactive analog 4alpha-phorbol 12,13-didecanoate had no effect. Western blot analysis detected an inward rectifying K+ channel (Kir6.2) immunoreactive protein at 56 kDa and PKC-delta in the IMM. These data provide the first characterization of the human cardiac mitoKATP channel and its regulation by PKC(s) in IMM. This local PKC control mechanism may represent an alternative pathway to that proposed previously for cytosolic PKC during ischemic/pharmacological preconditioning.
Collapse
Affiliation(s)
- Ming Tao Jiang
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Zhu YH, Wang X. Overexpression of heat-shock protein 20 in rat heart myogenic cells confers protection against simulated ischemia/reperfusion injury. Acta Pharmacol Sin 2005; 26:1076-80. [PMID: 16115374 DOI: 10.1111/j.1745-7254.2005.00137.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM To explore whether overexpression of the small heat shock protein HSP20 in rat cardiomyocytes protects against simulated ischemia/reperfusion (SI/R) injury. METHODS Recombinant adenovirus expressing HSP20 was used to infect rat H9c2 cardiomyocytes at high efficiency, as assessed by green fluorescent protein. H9c2 cells were subjected to SI/R stress; survival was estimated through assessment of lactate dehydrogenase and cell apoptosis through caspase-3 activity. RESULTS Overexpression of HSP20 decreased lactate dehydrogenase release by 21.5% and caspase-3 activity by 58.8%. Pretreatment with the protein kinase C inhibitor Ro-31-8220 (0.1 micromol/L) for 30 min before SI/R canceled the protective effect of HSP20. The selective mitochondrial K+ATP channel inhibitor 5-hydroxydecanoate (100 micromol/L) had a similar effect. However, the non-selective K+ATP channel inhibitor glibenclamide (100 micromol/L) had no significant effect. CONCLUSION These data indicate that the protective effect of HSP20 in vitro is primarily due to reduced necrotic and apoptotic death of cardiomyocytes, possibly via the protein kinase C/mitochondrial K+ATP pathway.
Collapse
Affiliation(s)
- Yan-hui Zhu
- Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Biology, Ministry of Education, Peking University, Beijing 100083, China
| | | |
Collapse
|
47
|
Liang HW, Xia Q, Bruce IC. Reactive oxygen species mediate the neuroprotection conferred by a mitochondrial ATP-sensitive potassium channel opener during ischemia in the rat hippocampal slice. Brain Res 2005; 1042:169-75. [PMID: 15854588 DOI: 10.1016/j.brainres.2005.02.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2004] [Revised: 02/04/2005] [Accepted: 02/12/2005] [Indexed: 11/25/2022]
Abstract
Reactive oxygen species (ROS) are known to mediate the protection conferred by the opening of mitochondrial ATP-sensitive potassium channels (mitoK(ATP)) during ischemia in heart, but this has not been demonstrated in brain. The present study examined whether ROS mediate the neuroprotection conferred by a mitoK(ATP) opener during ischemia in rat hippocampal slices. Ischemia was simulated by oxygen and glucose deprivation. The direct current potential and population spike were recorded in the stratum pyramidale of the CA1 region, and lactate dehydrogenase (LDH) efflux into the medium was assayed. ROS generation was measured spectrophotofluorometrically. Pretreatment of slices with diazoxide (DIA, 300 microM), a mitoK(ATP) opener, (i) prolonged the latency to ischemic depolarization and decreased its amplitude, (ii) delayed the onset of population spike disappearance and enhanced its recovery after reperfusion, (iii) decreased LDH efflux and (iv) increased ROS levels. The effects induced by DIA were attenuated by 5-hydroxydecanoic acid (200 microM), a mitoK(ATP) blocker. Pretreatment with N-2-mercaptopropionyl glycine (MPG, 500 microM), a ROS scavenger, also abrogated the effects induced by DIA, while treatment with MPG alone had no effect during normoxia and ischemia. These results indicate that ROS participate in the neuroprotection conferred by a mitoK(ATP) opener during ischemia.
Collapse
Affiliation(s)
- Hua-Wei Liang
- Department of Physiology, Zhejiang University School of Medicine, 353# Yan-an Road, Hangzhou 310031, China
| | | | | |
Collapse
|
48
|
Zhuo ML, Huang Y, Liu DP, Liang CC. KATP channel: relation with cell metabolism and role in the cardiovascular system. Int J Biochem Cell Biol 2005; 37:751-64. [PMID: 15694835 DOI: 10.1016/j.biocel.2004.10.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Indexed: 12/21/2022]
Abstract
ATP-sensitive potassium channel (K(ATP)) is one kind of inwardly rectifying channel composed of two kinds of subunits: the pore forming subunits and the regulatory subunits. K(ATP) channels exist in the sarcolemmal, mitochondrial and nuclear membranes of various tissues. Cell metabolism regulates K(ATP) gene expression and metabolism products regulate the channel by direct interactions, while K(ATP) controls membrane potentials and regulate cell activities including energy metabolism, apoptosis and gene expression. K(ATP) channels from different cell organelles are linked by some signal molecules and they can respond to common stimulation in a coordinate way. In the cardiovascular system K(ATP) has important functions. The most prominent is that opening of this channel can protect cardiac myocytes against ischemic injuries. The sarcolemmal K(ATP) may provide a basic protection against ischemia by energy sparing, while both the sarcolemmal K(ATP) and mitochondrial K(ATP) channels are necessary for the ischemia preconditioning. K(ATP) channels also have important functions including homeostasis maintenance and vascular tone regulation under physiological conditions. Further elucidation of the role of K(ATP) in the cardiovascular system will help us to regulate cell metabolism or prevent damage caused by abnormal channel functions.
Collapse
Affiliation(s)
- Ming-Lei Zhuo
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Dong Dan San Tiao 5, Beijing 100005, PR China
| | | | | | | |
Collapse
|
49
|
Wang W, Jia L, Wang T, Sun W, Wu S, Wang X. Endogenous Calcitonin Gene-related Peptide Protects Human Alveolar Epithelial Cells through Protein Kinase Cϵ and Heat Shock Protein. J Biol Chem 2005; 280:20325-30. [PMID: 15784626 DOI: 10.1074/jbc.m413864200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The intracellular mechanisms of ischemic preconditioning (PC) in preventing lung dysfunction following transplantation, shock, and trauma remain poorly understood. Previously, we have shown that alveolar epithelial cells secrete calcitonin gene-related peptide (CGRP) under inflammatory stress. Using a hypoxia/reoxygenation (H/R) and PC model, we found that CGRP was also secreted from human type II alveolar epithelial cells (A549) after PC. The locally released CGRP interacted with its receptor on the membrane of A549 cells and elicited downstream signals mediating the PC effect, because hCGRP(8-37), a specific CGRP receptor antagonist, attenuated the protective effect of PC. Pre-inhibition of CGRP protein synthesis by small interfering RNA exacerbated (but overexpression of the CGRP gene ameliorated) H/R-induced cell death, which supports the autocrine effect of CGRP on A549 cells. Exogenous bioactive CGRP mimicked the beneficial effect of PC and up-regulated the expression of heat shock protein 70 (HSP70), which might act as the end effector to maintain cell viability. These effects were sensitive to hCGRP(8-37), calphostin C (a protein kinase C (PKC) inhibitor), and 5-hydroxydecanoic acid (a mitochondrial K(+)(ATP) channel blocker) but were insensitive to protein kinase A blockers. Moreover, CGRP induced the membrane translocation of PKCepsilon. PKCV1-2 (a cell-permeable inhibitory peptide of PKCepsilon) effectively abolished CGRP-induced HSP70 expression and cell protection. Therefore, PC induces CGRP secretion from human alveolar epithelial cells, and the locally released CGRP acts back on these cells, protecting them from H/R injury. The post-receptor signaling of CGRP is through PKCepsilon-dependent expression of HSP70.
Collapse
Affiliation(s)
- Wang Wang
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100083, China
| | | | | | | | | | | |
Collapse
|
50
|
Mironov SL, Hartelt N, Ivannikov MV. Mitochondrial K(ATP) channels in respiratory neurons and their role in the hypoxic facilitation of rhythmic activity. Brain Res 2005; 1033:20-7. [PMID: 15680335 DOI: 10.1016/j.brainres.2004.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2004] [Indexed: 01/25/2023]
Abstract
Hypoxia is damaging in neurons, but it can also produce beneficial effects by consolidating the activity of neural networks such as facilitation of respiratory activity [T.L. Baker-Herman, D.D. Fuller, R.W. Bavis, A.G. Zabka, F.J. Golder, N.J. Doperalski, R.A. Johnson, J.J. Watters, G.S. Mitchell, Nature Neuroscience 7 (2004) 48-55; J.L. Feldman, G.S. Mitchell, E.E. Nattie, Ann. Rev. Neurosci. 26 (2003) 239-266; D.M. Blitz, J.M. Ramirez, J. Neurophysiol. 87 (2002) 2964-2971]. The underlying mechanisms are unknown, and we hypothesized they may be similar to ischemic preconditioning in the heart, involving mitochondrial K(ATP) (mK(ATP)) channels. By measuring the mitochondrial potential (Psi(m)) and Ca2+ ([Ca2+]m) in neurons of pre-Botzinger complex (pBC), we examined the functional expression of mK(ATP) channels in the respiratory network. The opener of mK(ATP) channels diazoxide decreased Psi(m) and [Ca2+]m both in pBC neurons and in isolated immobilized mitochondria. 5-Hydroxydecanoate (5-HD), the blocker of mK(ATP) channels, increased both Psi(m) and [Ca2+]m. Phorbol 12-myristate-13-acetate (PMA) mimicked the effects of diazoxide. Protein kinase C (PKC) was stimulated during hypoxia that occurred mostly at the mitochondria. Brief hypoxia induced facilitation of the respiratory activity, which was prevented after blockade of mK(ATP) channels with 5-HD and PKC with staurosporine. Diazoxide potentiated the motor output and subsequent application of hypoxia was ineffective. We propose that a PKC-induced stimulation of K(ATP) channels in the mitochondria of respiratory neurons is responsible for the hypoxic facilitation of rhythmic activity.
Collapse
Affiliation(s)
- S L Mironov
- Department Neuro- and Sensory Physiology, Georg August University, Humboldtallee 23, 37073 Göttingen, Germany.
| | | | | |
Collapse
|