101
|
Facundo HTF, de Paula JG, Kowaltowski AJ. Mitochondrial ATP-Sensitive K+ Channels Prevent Oxidative Stress, Permeability Transition and Cell Death. J Bioenerg Biomembr 2005; 37:75-82. [PMID: 15906152 DOI: 10.1007/s10863-005-4130-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2005] [Accepted: 01/17/2005] [Indexed: 10/25/2022]
Abstract
Ischemia followed by reperfusion results in impairment of cellular and mitochondrial functionality due to opening of mitochondrial permeability transition pores. On the other hand, activation of mitochondrial ATP-sensitive K(+) channels (mitoK(ATP)) protects the heart against ischemic damage. This study examined the effects of mitoK(ATP) and mitochondrial permeability transition on isolated rat heart mitochondria and cardiac cells submitted to simulated ischemia and reperfusion (cyanide/aglycemia). Both mitoK(ATP) opening, using diazoxide, and the prevention of mitochondrial permeability transition, using cyclosporin A, protected against cellular damage, without additive effects. MitoK(ATP) opening in isolated rat heart mitochondria slightly decreased Ca(2+) uptake and prevented mitochondrial reactive oxygen species production, most notably in the presence of added Ca(2+). In ischemic cells, diazoxide decreased ROS generation during cyanide/aglycemia while cyclosporin A prevented oxidative stress only during simulated reperfusion. Collectively, these studies indicate that opening mitoK(ATP) prevents cellular death under conditions of ischemia/reperfusion by decreasing mitochondrial reactive oxygen species release secondary to Ca(2+) uptake, inhibiting mitochondrial permeability transition.
Collapse
Affiliation(s)
- Heberty T F Facundo
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-900, Brazil
| | | | | |
Collapse
|
102
|
Korge P, Honda HM, Weiss JN. K+-dependent regulation of matrix volume improves mitochondrial function under conditions mimicking ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2005; 289:H66-77. [PMID: 15764674 DOI: 10.1152/ajpheart.01296.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
To delineate the role of mitochondrial K+ fluxes in cardioprotection, we investigated the effect of extramitochondrial K+ on the ability of mitochondria to support membrane potential (DeltaPsi), regulate matrix volume, consume oxygen, and phosphorylate ADP under conditions mimicking key elements of ischemia-reperfusion. Isolated energized mitochondria responded to ADP addition with depolarization, increased O2 consumption, and matrix shrinkage. The time required for full recovery of DeltaPsi, signaling the completion of ADP phosphorylation, was used to evaluate the rate of ATP synthesis during repeated ADP pulses. In mitochondria with a decreased ability to support DeltaPsi, the rate of ADP phosphorylation was significantly improved by extramitochondrial K+ > Na+ > Li+, especially at higher buffer osmolarity, which promotes matrix shrinkage. K+-induced improvement in DeltaPsi recovery after ADP pulses was accompanied by more rapid and complete matrix volume recovery and enhanced O2 consumption. Manipulations expected to affect matrix swelling by regulating K+ fluxes or water distribution indicate that matrix volume regulation by external factors becomes increasingly important in mitochondria with decreased ability to support DeltaPsi in the face of a high ADP load. Under these conditions, opening of K+ influx pathways improved mitochondrial function and delayed failure. This may be an important factor in the mechanism of diaxozide-induced cardioprotection.
Collapse
Affiliation(s)
- Paavo Korge
- Cardiovascular Research Laboratory, 3645 MRL Bldg., 675 Charles Young Dr. S., David Geffen School of Medicine, Univ. of California, Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|
103
|
Busija DW, Lacza Z, Rajapakse N, Shimizu K, Kis B, Bari F, Domoki F, Horiguchi T. Targeting mitochondrial ATP-sensitive potassium channels--a novel approach to neuroprotection. ACTA ACUST UNITED AC 2005; 46:282-94. [PMID: 15571770 DOI: 10.1016/j.brainresrev.2004.06.011] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2004] [Indexed: 10/26/2022]
Abstract
Mitochondrial responses to ischemic stress play an important role in necrosis and apoptosis of brain cells. Recent studies using several different experimental preparations have shown that activation of ATP-sensitive potassium channels in mitochondria (mitoK(ATP) channels) is able to protect neurons and astroglia against injury and death. Thus, targeting of mitoK(ATP) channels appears to be a novel approach to neuroprotection. However, little is known about the mechanisms involved. The purpose of this review is to detail the current state of knowledge about this important, emerging area of investigation, and to provide suggestions for future studies.
Collapse
Affiliation(s)
- David W Busija
- Department of Physiology and Pharmacology, and Center for Investigative Neuroscience, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157-1010, USA.
| | | | | | | | | | | | | | | |
Collapse
|
104
|
Ardehali H, O'Rourke B. Mitochondrial K(ATP) channels in cell survival and death. J Mol Cell Cardiol 2005; 39:7-16. [PMID: 15978901 PMCID: PMC2692534 DOI: 10.1016/j.yjmcc.2004.12.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Revised: 12/09/2004] [Accepted: 12/10/2004] [Indexed: 01/27/2023]
Abstract
Since the discovery of the mitochondrial ATP-sensitive potassium channel (mitoK(ATP)) more than 13 years ago, it has been implicated in the processes of ischemic preconditioning (IPC), apoptosis and mitochondrial matrix swelling. Different approaches have been employed to characterize the pharmacological profile of the channel, and these studies strongly suggest that cellular protection well correlates with the opening of mitoK(ATP). However, there are many questions regarding mitoK(ATP) that remain to be answered. These include the very existence of mitoK(ATP) itself, its degree of importance in the process of IPC, its response to different pharmacological agents, and how its activation leads to the process of IPC and protection against cell death. Recent findings suggest that mitoK(ATP) may be a complex of multiple mitochondrial proteins, including some which have been suggested to be components of the mitochondrial permeability transition pore. However, the identity of the pore-forming unit of the channel and the details of the interactions between these proteins remain unclear. In this review, we attempt to highlight the recent advances in the physiological role of mitoK(ATP) and discuss the controversies and unanswered questions.
Collapse
Affiliation(s)
| | - Brian O'Rourke
- Corresponding author. Tel.: +1-410-614-0034; fax: +1-410-955-7953. E-mail address: (B. O'Rourke)
| |
Collapse
|
105
|
Marín-García J. Cellular and molecular events in ischemic preconditioning: potential therapeutic applications in cardioprotection. Future Cardiol 2005; 1:111-22. [DOI: 10.1517/14796678.1.1.111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Cardioprotection is a mechanism of guarding the heart from damage secondary to different insults including ischaemia, ischaemia/reperfusion, chemical, metabolic and physical stressors. Ischemic preconditioning, by single or multiple brief periods of ischaemia, protects the heart against a more prolonged ischemic insult (index ischaemia). Understanding the cellular, molecular and biochemical events occurring in cardioprotection will allow the development of new interventions to improve the outcome of patients with myocardial diseases. Most of the present experience with cardioprotection has been obtained from studies in young and middle-aged animals, and cells. In the future, cardioprotection research should be carried out mainly in the aging or senescent heart since this will be most relevant to humans. With aging, the heart has a decreased capacity to tolerate and respond to various forms of stress, and the likelihood of myocardial ischaemia and cardiac dysfunction increases.
Collapse
Affiliation(s)
- José Marín-García
- RWJ. Medical School, Department of Physiology & Biophysics, The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Ave., Highland Park, NJ 08904, USATel.: Fax:
| |
Collapse
|
106
|
Simpson D, Wellington K. Nicorandil: a review of its use in the management of stable angina pectoris, including high-risk patients. Drugs 2004; 64:1941-55. [PMID: 15329045 DOI: 10.2165/00003495-200464170-00012] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nicorandil (Adancor, Angicor, Dancor, Nikoril [Europe], Ikorel [Europe and Oceania], Sigmart [Japan, Korea and Taiwan]) is an adenosine triphosphate (ATP)-sensitive potassium (KATP) channel agonist with nitrate-like properties used in the management of stable angina pectoris. With well established monotherapeutic antianginal activity and a beneficial effect (when added to optimal antianginal therapy) on clinical outcomes in high-risk patients with stable angina, twice-daily oral nicorandil is a useful alternative or addition to other antianginal therapy.
Collapse
Affiliation(s)
- Dene Simpson
- Adis International Limited, Auckland, New Zealand.
| | | |
Collapse
|
107
|
Domoki F, Bari F, Nagy K, Busija DW, Siklós L. Diazoxide prevents mitochondrial swelling and Ca2+ accumulation in CA1 pyramidal cells after cerebral ischemia in newborn pigs. Brain Res 2004; 1019:97-104. [PMID: 15306243 DOI: 10.1016/j.brainres.2004.05.088] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2004] [Indexed: 01/05/2023]
Abstract
Diazoxide (DIAZ), an opener of mitochondrial ATP-sensitive K(+) channels (mK(ATP)), protects neurons against hypoxic/ischemic stress in vivo, however, direct evidence showing mitochondrial effects of DIAZ in postischemic neurons is lacking. We investigated if DIAZ affects mitochondrial alterations after global ischemia/reperfusion (I/R) in CA1 pyramidal neurons by using oxalate-pyroantimonate electron cytochemistry. Anesthetized piglets were either non-treated, or treated with DIAZ (3 mg/kg, iv), I/R, DIAZ+I/R, or 5-hydroxy-decanoate (5HD)+DIAZ+I/R (n=6, 6, 11, 5, 7, respectively). Ischemia (10 min) was induced by intracranial pressure (ICP) elevation. After 5-30 min of reperfusion, the brains were fixed for ultrastructural studies. Relative volumes of Ca(2+)-containing deposits and mitochondria in CA1 pyramidal cells were determined by point counting on electron micrographs. I/R resulted in maximal increases in mitochondrial volume (from 7.14+/-0.63% to 9.74+/-0.57%*), and Ca(2+) levels (from 5.86+/-1.11% to 11.39+/-1.35%*; mean+/-S.E.M., *p<0.05) at 10-15-min reperfusion time. In this interval, pretreatment with DIAZ virtually abolished mitochondrial swelling (6.88+/-0.49%) and Ca(2+) accumulation (5.15+/-0.82%) evoked by I/R. The protective effect of DIAZ was reduced by 5HD, an inhibitor of mK(ATP), resulting in a calcium accumulation similar to that after IR (10.44+/-1.98%). Thus, DIAZ might preserve mitochondrial integrity in CA1 pyramidal cells after I/R, at least in part mediated by mK(ATP).
Collapse
Affiliation(s)
- Ferenc Domoki
- Department of Physiology, Faculty of Medicine, University of Szeged, Szeged, Dóm tér 10. H-6720, Hungary.
| | | | | | | | | |
Collapse
|
108
|
Wakahara N, Katoh H, Yaguchi Y, Uehara A, Satoh H, Terada H, Fujise Y, Hayashi H. Difference in the cardioprotective mechanisms between ischemic preconditioning and pharmacological preconditioning by diazoxide in rat hearts. Circ J 2004; 68:156-62. [PMID: 14745152 DOI: 10.1253/circj.68.156] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recent studies have implicated the opening of mitochondrial K(ATP) (mitoK(ATP)) channels and the production of reactive oxygen species (ROS) in the cardioprotective mechanism of ischemic preconditioning (IPC). METHODS AND RESULTS The involvement of mitoK(ATP) channels and ROS in the cardioprotective effects of both IPC and the mitoK(ATP) channel opener diazoxide (DZ) was investigated in ischemic/reperfused rat hearts. The effects of IPC and DZ on myocardial high-energy phosphate concentrations and intracellular pH (pH(i)) were also examined using (31)P nuclear magnetic resonance spectroscopy. Although both the mitoK(ATP) channel inhibitor 5-hydroxydecanoate and the antioxidant N-acetylcysteine abolished the postischemic recovery of contractile function by DZ, neither of them inhibited that by IPC. IPC attenuated the decline in pHi during ischemia, but DZ did not (6.28+/-0.04 in IPC, p<0.05, and 6.02+/-0.05 in DZ vs 6.02 +/-0.06 in control hearts). DZ, but not IPC, reduced the decrease in ATP levels during ischemia (ATP levels at 20-min ischemia: 26.3+/-3.4% of initial value in DZ, p<0.05, and 8.1+/-3.0% in IPC vs 15.1+/-1.3% in control hearts). CONCLUSIONS These results suggest that DZ-induced cardioprotection is related to ROS production and reduced ATP degradation during ischemia, whereas attenuated acidification during ischemia is involved in IPC-induced cardioprotection, which is not mediated through mitoK(ATP) channel opening or ROS production.
Collapse
Affiliation(s)
- Nobuyuki Wakahara
- Division of Cardiology, Department of Internal Medicine III, Hamamatsu University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Kopustinskiene DM, Pollesello P, Saris NEL. Potassium-specific effects of levosimendan on heart mitochondria. Biochem Pharmacol 2004; 68:807-12. [PMID: 15294443 DOI: 10.1016/j.bcp.2004.05.018] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Accepted: 05/11/2004] [Indexed: 11/22/2022]
Abstract
In this study, we evaluated levosimendan, a new drug developed for the treatment of acute and decompensated heart failure, as a potential activator of ATP-sensitive potassium flux to the matrix of cardiac mitochondria. We estimated the KATP channel openers-induced increase in mitochondrial inner membrane permeability for potassium by registering changes in membrane potential of heart mitochondria, oxidizing endogenous substrates. We compared the effect of levosimendan with the effects of the known KATP channel openers diazoxide and pinacidil. Levosimendan (1 microM) accelerated potassium-specific DeltaPsi decrease by 0.15%/s, whereas 50 microM diazoxide by 0.10%/s, and 50 microM pinacidil by 0.08%/s, respectively. These results were confirmed by swelling experiments of non-respiring mitochondria in potassium nitrate medium. We found that levosimendan with an EC50 of 0.83 +/- 0.24 microM activates potassium flux to the mitochondrial matrix. This effect is discussed as a possible explanation of the anti-ischemic action of levosimendan.
Collapse
|
110
|
Abstract
BACKGROUND There is increasing evidence documenting the capacity of myocardial cells exposed to a variety of insults to mount a cardioprotective response. Although this cardioprotection has been most well characterized with respect to ischemic preconditioning, other chemical and metabolic stressors have been shown to share features of the ischemic preconditioning model, including the involvement of mitochondria in the triggering, signaling, and mediation of the cardioprotective response. METHODS In this article, we review the evidence showing that mitochondria play a critical role in cardioprotection from multiple (often interrelated) standpoints: its primary function in producing the cellular bioenergetic supply, its control over events in apoptosis, its contribution to myocardial signal transducing processes, and its role in producing reactive oxidative species and in providing an appropriate antioxidant response to a variety of cellular insults. CONCLUSIONS Although our understanding of cytoprotection has increased substantially within the last few years, the mechanisms mediating mitochondrial resistance to insults leading to cardiac protection remain to be fully delineated, and represents a significant approach in the clinical treatment of heart disease.
Collapse
Affiliation(s)
- José Marín-García
- Molecular Cardiology and Neuromuscular Institute, 75 Raritan Avenue, Highland Park, NJ 08904, USA
| | | |
Collapse
|
111
|
McLeod CJ, Jeyabalan AP, Minners JO, Clevenger R, Hoyt RF, Sack MN. Delayed ischemic preconditioning activates nuclear-encoded electron-transfer-chain gene expression in parallel with enhanced postanoxic mitochondrial respiratory recovery. Circulation 2004; 110:534-9. [PMID: 15277332 DOI: 10.1161/01.cir.0000136997.53612.6c] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Delayed ischemic preconditioning promotes cardioprotection via genomic reprogramming. We hypothesize that molecular regulation of mitochondrial energetics is integral to this cardioprotective program. METHODS AND RESULTS Preconditioning was induced by use of 3 episodes of 3-minute coronary artery occlusion separated by 5 minutes of reperfusion. Twenty-four hours later, infarct size was reduced by 58% after preconditioning compared with sham-operated controls (P<0.001). Cardiac mitochondria were isolated from sham and preconditioned rat hearts. Mitochondrial respiration and ATP production were similar between the groups; however, preconditioned mitochondria exhibit modest hyperpolarization of the inner mitochondrial membrane potential (> or =22% versus control, P<0.001). After 35-minute anoxia and reoxygenation, preconditioned mitochondria demonstrated a 191+/-12% improvement in ADP-sensitive respiration (P=0.002) with preservation of electron-transfer-chain (ETC) activity versus controls. This augmented mitochondrial recovery was eradicated when preconditioning was abolished by the antioxidant 2-mercaptopropionyl glycine (2-MPG). These biochemical modulations appear to be regulated at the genomic level in that the expression of genes encoding rate-controlling complexes in the ETC was significantly upregulated in preconditioned myocardium, with a concordant induction of steady-state protein levels of cytochrome oxidase, cytochrome c, and adenine nucleotide translocase-1. 2-MPG abolished preconditioning induction of these transcripts. Moreover, transcripts of nuclear regulatory peptides known to orchestrate mitochondrial biogenesis, nuclear respiratory factor-1 and peroxisome-proliferator-activated receptor gamma coactivator 1alpha, were significantly induced in preconditioned myocardium. CONCLUSIONS Delayed preconditioned mitochondria display increased tolerance against anoxia-reoxygenation in association with modifications in mitochondrial bioenergetics, with concordant genomic induction of a mitochondrial energetic gene regulatory program. This program appears to be mediated by reactive oxygen species signaling.
Collapse
Affiliation(s)
- Christopher J McLeod
- Cardiovascular Branch, NHLBI, National Institutes of Health, Bethesda, Md 20892-1650, USA
| | | | | | | | | | | |
Collapse
|
112
|
Nagy K, Kis B, Rajapakse NC, Bari F, Busija DW. Diazoxide preconditioning protects against neuronal cell death by attenuation of oxidative stress upon glutamate stimulation. J Neurosci Res 2004; 76:697-704. [PMID: 15139028 DOI: 10.1002/jnr.20120] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We examined the effects of diazoxide, the putative mitochondrial adenosine triphosphate-sensitive potassium (mitoK(ATP)) channel opener, against glutamate excitotoxicity in primary cultures of rat cortical neurons. Cells were treated with diazoxide for 24 hr and then exposed to 200 microM glutamate. Cell viability was measured 24 hr after glutamate exposure. We found that treatment 24 hr before glutamate exposure with 250 and 500 microM diazoxide but not with another mitoK(ATP) channel opener, nicorandil, increased neuronal viability from 54 +/- 2% to 84 +/- 2% and 92 +/- 3%, respectively (n = 25-40). These effects were not inhibited by the putative mitoK(ATP) channel blocker 5-hydroxydecanoic acid. Diazoxide application increased production of reactive oxygen species (ROS) and coapplication of M40401, a superoxide dismutase mimetic, prevented delayed preconditioning. The 24 hr preconditioned neurons showed significantly reduced ROS production upon glutamate stimulation compared to that in untreated cells. These results suggest that diazoxide induces delayed preconditioning in cultured cortical neurons via increased ROS production and attenuation of oxidative stress upon glutamate stimulation.
Collapse
Affiliation(s)
- Krisztina Nagy
- Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | |
Collapse
|
113
|
Kis B, Nagy K, Snipes JA, Rajapakse NC, Horiguchi T, Grover GJ, Busija DW. The mitochondrial K(ATP) channel opener BMS-191095 induces neuronal preconditioning. Neuroreport 2004; 15:345-9. [PMID: 15076766 DOI: 10.1097/00001756-200402090-00027] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BMS-191095, reportedly a selective mitoK(ATP) channel opener which is free from the known side effects of the prototype mitoK(ATP) channel opener diazoxide, induced acute and delayed preconditioning against glutamate excitotoxicity and delayed preconditioning against oxygen-glucose deprivation in primary cultures of rat cortical neurons. BMS-191095 dose dependently depolarized the mitochondria, increased the phosphorylation of PKC isoforms, but had no detectable effects on the activation of MAP kinases and did not influence the expressions of HSP70 and Mn-SOD. In BMS-191095-preconditioned neurons the glutamate-induced free-radical production was abolished. Our data give the first evidence that selective opening of mitoK(ATP) channels with BMS-191095 leads to remarkable neuroprotection via mechanisms that involve mitochondrial depolarization, PKC activation and attenuated free radical production during neuronal stress.
Collapse
Affiliation(s)
- Bela Kis
- Department of Physiology and Pharmacology, Wake Forest University, School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | | | | | | | | | | | | |
Collapse
|
114
|
Abstract
Twenty years after the discovery of sarcolemmal ATP-sensitive K+ channels and 12 years after the discovery of mitochondrial K(ATP) (mitoK(ATP)) channels, progress has been remarkable, but many questions remain. In the case of the former, detailed structural information is available, and it is well accepted that the channel couples bioenergetics to cellular electrical excitability; however, in the heart, a clear physiological or pathophysiological role has yet to be defined. For mitoK(ATP), structural information is lacking, but there is abundant evidence linking the opening of the channel to protection against ischemia-reperfusion injury or apoptosis. This review updates recent progress in understanding the physiological role of mitoK(ATP) and highlights outstanding questions and controversies, with the intent of stimulating additional investigation on this topic.
Collapse
Affiliation(s)
- Brian O'Rourke
- Institute of Molecular Cardiobiology, Johns Hopkins University, 720 Rutland Ave, 844 Ross Bldg, Baltimore, Md 21205-2195, USA.
| |
Collapse
|
115
|
Budas GR, Jovanovic S, Crawford RM, Jovanovic A. Hypoxia-induced preconditioning in adult stimulated cardiomyocytes is mediated by the opening and trafficking of sarcolemmal KATP channels. FASEB J 2004; 18:1046-8. [PMID: 15084521 PMCID: PMC2128706 DOI: 10.1096/fj.04-1602fje] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The opening of sarcolemmal and mitochondrial ATP-sensitive K(+) (KATP) channels in the heart is believed to mediate ischemic preconditioning, a phenomenon whereby brief periods of ischemia/reperfusion protect the heart against myocardial infarction. Here, we have applied digital epifluorescent microscopy, immunoprecipitation and Western blotting, perforated patch clamp electrophysiology, and immunofluorescence/laser confocal microscopy to examine the involvement of KATP channels in cardioprotection afforded by preconditioning. We have shown that adult, stimulated-to-beat, guinea-pig cardiomyocytes survived in sustained hypoxia for approximately 17 min. An episode of 5-min-long hypoxia/5-min-long reoxygenation before sustained hypoxia dramatically increased the duration of cellular survival. Experiments with different antagonists of KATP channels, applied at different times during the experimental protocol, suggested that the opening of sarcolemmal KATP channels at the beginning of sustained hypoxia mediate preconditioning. This conclusion was supported by perforated patch clamp experiments that revealed activation of sarcolemmal KATP channels by preconditioning. Immunoprecipitation and Western blotting as well as immunofluorescence and laser confocal microscopy showed that the preconditioning is associated with the increase in KATP channel proteins in sarcolemma. Inhibition of trafficking of KATP channel subunits prevented preconditioning without affecting sensitivity of cardiomyocytes to hypoxia in the absence of preconditioning. We conclude that the preconditioning is mediated by the activation and trafficking of sarcolemmal KATP channels.
Collapse
Affiliation(s)
- Grant R Budas
- Maternal and Child Health Sciences, Tayside Institute of Child Health, Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, UK
| | | | | | | |
Collapse
|
116
|
Hausenloy DJ, Yellon DM, Mani-Babu S, Duchen MR. Preconditioning protects by inhibiting the mitochondrial permeability transition. Am J Physiol Heart Circ Physiol 2004; 287:H841-9. [PMID: 15072953 DOI: 10.1152/ajpheart.00678.2003] [Citation(s) in RCA: 171] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitochondrial permeability transition (mPT) is a crucial event in the progression to cell death in the setting of ischemia-reperfusion. We have used a model system in which mPT can be reliably and reproducibly induced to test the hypothesis that the profound protection associated with the phenomenon of myocardial preconditioning is mediated by suppression of the mPT. Adult rat myocytes were loaded with the fluorescent probe tetramethylrhodamine methyl ester, which generates oxidative stress on laser illumination, thus inducing the mPT (indicated by collapse of the mitochondrial membrane potential) and ATP depletion, seen as rigor contracture. The known inhibitors of the mPT, cyclosporin A (0.2 microM) and N-methyl-4-valine-cyclosporin A (0.4 microM), increased the time taken to induce the mPT by 1.8- and 2.9-fold, respectively, compared with control (P < 0.001) and rigor contracture by 1.5-fold compared with control (P < 0.001). Hypoxic preconditioning (HP) and pharmacological preconditioning, using diazoxide (30 microM) or nicorandil (100 microM), also increased the time taken to induce the mPT by 2.0-, 2.1-, and 1.5-fold, respectively (P < 0.001), and rigor contracture by 1.9-, 1.7-, and 1.5-fold, respectively, compared with control (P < 0.001). Effects of HP, diazoxide, and nicorandil were abolished in the presence of mitochondrial ATP-sensitive K(+) (K(ATP)) channel blockers glibenclamide (10 microM) and 5-hydroxydecanoate (100 microM) but were maintained in the presence of the sarcolemmal K(ATP) channel blocker HMR-1098 (10 microM). In conclusion, preconditioning protects the myocardium by reducing the probability of the mPT, which normally occurs during ischemia-reperfusion in response to oxidative stress.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Institute and Centre for Cardiology, University College London Hospitals and Medical School, Grafton Way, London WC1E 6DB, UK
| | | | | | | |
Collapse
|
117
|
Abstract
Ischemic preconditioning (IPC) is a most powerful endogenous mechanism for myocardial protection against ischemia/reperfusion injury. It is now apparent that reactive oxygen species (ROS) generated in the mitochondrial respiratory chain act as a trigger of IPC. ROS mediate signal transduction in the early phase of IPC through the posttranslational modification of redox-sensitive proteins. ROS-mediated activation of Src tyrosine kinases serves a scaffold for interaction of proteins recruited by G protein-coupled receptors and growth factor receptors that is necessary for amplification of cardioprotective signal transduction. Protein kinase C (PKC) plays a central role in this signaling cascade. A crucial target of PKC is the mitochondrial ATP-sensitive potassium channel, which acts as a trigger and a mediator of IPC. Mitogen-activated protein (MAP) kinases (extracellular signal-regulated kinase, p38 MAP kinase, and c-Jun NH(2)-terminal kinase) are thought to exist downstream of the Src-PKC signaling module, although the role of MAP kinases in IPC remains undetermined. The late phase of IPC is mediated by cardioprotective gene expression. This mechanism involves redox-sensitive activation of transcription factors through PKC and tyrosine kinase signal transduction pathways that are in common with the early phase of IPC. The effector proteins then act against myocardial necrosis and stunning presumably through alleviation of oxidative stress and Ca(2+) overload. Elucidation of IPC-mediated complex signaling processes will help in the development of more effective pharmacological approaches for prevention of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Hajime Otani
- Department of Thoracic and Cardiovascular Surgery, Kansai Medical University, Moriguchi City, Osaka 570, Japan.
| |
Collapse
|
118
|
Teshima Y, Akao M, Baumgartner WA, Marbán E. Nicorandil prevents oxidative stress-induced apoptosis in neurons by activating mitochondrial ATP-sensitive potassium channels. Brain Res 2004; 990:45-50. [PMID: 14568328 DOI: 10.1016/s0006-8993(03)03383-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nicorandil, a clinically useful drug for the treatment of ischemic heart disease, has an anti-apoptotic effect in cardiomyocytes, and activation of mitochondrial ATP-sensitive potassium (mitoKATP) channels underlies this effect. Recently, several studies showed that nicorandil reduced brain injury in animal models of brain ischemia. Based on these facts, we hypothesized that nicorandil may have anti-apoptotic effects in neurons mediated by mitoKATP channels. We investigated the effect of nicorandil on apoptosis induced by oxidative stress using cultured cerebellar granule neurons. Nicorandil (100 micromol/l) significantly suppressed the number of cells with TUNEL-positive nuclei and the increase in caspase-3 activity induced by 20 micromol/l H2O2. An indicator dye for mitochondrial inner membrane potential (DeltaPsim) revealed that nicorandil prevented the loss of DeltaPsim induced by H2O2 in a concentration-dependent manner. These effects were abolished by 5-hydroxydecanoate (5HD; 500 micromol/l), a mitoKATP channel blocker. The present results showed that nicorandil has anti-apoptotic effects in neurons, at least in part, by preserving DeltaPsim.
Collapse
Affiliation(s)
- Yasushi Teshima
- Institute of Molecular Cardiobiology, The Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
119
|
Kuniyasu A, Kaneko K, Kawahara K, Nakayama H. Molecular assembly and subcellular distribution of ATP-sensitive potassium channel proteins in rat hearts. FEBS Lett 2003; 552:259-63. [PMID: 14527696 DOI: 10.1016/s0014-5793(03)00936-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Cardiac ATP-sensitive K(+) (K(ATP)) channels are proposed to contribute to cardio-protection and ischemic preconditioning. Although mRNAs for all subunits of K(ATP) channels (Kir6.0 and sulfonylurea receptors SURs) were detected in hearts, subcellular localization of their proteins and the subunit combination are not well elucidated. We address these questions in rat hearts, using anti-peptide antibodies raised against each subunit. By immunoblot analysis, all of the subunits were detected in microsomal fractions including sarcolemmal membranes, while they were not detected in mitochondrial fractions at all. Immunoprecipitation and sucrose gradient sedimentation of the digitonin-solubilized microsomes indicated that Kir6.2 exclusively assembled with SUR2A. The molecular mass of the Kir6.2-SUR2A complex estimated by sucrose sedimentation was 1150 kDa, significantly larger than the calculated value for (Kir6.2)(4)-(SUR2A)(4), suggesting a potential formation of micellar complex with digitonin but no indication of hybrid channel formation under the conditions. These findings provide additional information on the structural and functional relationships of cardiac K(ATP) channel proteins involving subcellular localization and roles for cardioprotection and ischemic preconditioning.
Collapse
Affiliation(s)
- Akihiko Kuniyasu
- Department of Molecular Cell Function, Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, 5-1 Ohe-Honmachi, 862-0973 Kumamoto, Japan
| | | | | | | |
Collapse
|
120
|
Eigel BN, Gursahani H, Hadley RW. ROS are required for rapid reactivation of Na+/Ca2+ exchanger in hypoxic reoxygenated guinea pig ventricular myocytes. Am J Physiol Heart Circ Physiol 2003; 286:H955-63. [PMID: 14592940 DOI: 10.1152/ajpheart.00721.2003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cardiac Na(+)/Ca(2+) exchanger (NCX) contributes to cellular injury during hypoxia, as its altered function is largely responsible for a rise in cytosolic Ca(2+) concentration ([Ca(2+)](i)). In addition, the NCX in guinea pig ventricular myocytes undergoes profound inhibition during hypoxia and rapid reactivation during reoxygenation. The mechanisms underlying these changes in NCX activity are likely complex due to the participation of multiple inhibitory factors including altered cytosolic Na(+) concentration, pH, and ATP. Our main hypothesis is that oxidative stress is an essential trigger for rapid NCX reactivation in guinea pig ventricular myocytes and is thus a critical factor in determining the timing and magnitude of Ca(2+) overload. This hypothesis was evaluated in cardiac myocytes using fluorescent indicators to measure [Ca(2+)](i) and oxidative stress. An NCX antisense oligonucleotide was used to decrease NCX protein expression in some experiments. Our results indicate that NCX activity is profoundly inhibited in hypoxic guinea pig ventricular myocytes but is reactivated within 1-2 min of reoxygenation at a time of rising oxidative stress. We also found that several interventions to decrease oxidative stress including antioxidants and diazoxide prevented NCX reactivation and Ca(2+) overload during reoxygenation. Furthermore, application of exogenous H(2)O(2) was sufficient by itself to reactivate the NCX during sustained hypoxia and could reverse the suppression of reoxygenation-mediated NCX reactivation by diazoxide. These data suggest that elevated oxidative stress in reoxygenated guinea pig ventricular myocytes is required for rapid NCX reactivation, and thus reactivation should be viewed as an active process rather than being due to the simple decline of NCX inhibition.
Collapse
Affiliation(s)
- B N Eigel
- Dept. of Molecular and Biomedical Pharmacology, Univ. of Kentucky, College of Medicine, UKMC MS-375, Lexington, KY 40536-0298, USA.
| | | | | |
Collapse
|
121
|
Xiao XH, Allen DG. The cardioprotective effects of Na+/H+ exchange inhibition and mitochondrial KATP channel activation are additive in the isolated rat heart. Pflugers Arch 2003; 447:272-9. [PMID: 14534792 DOI: 10.1007/s00424-003-1183-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2003] [Revised: 07/31/2003] [Accepted: 09/05/2003] [Indexed: 01/22/2023]
Abstract
The mechanisms of recovery of the isolated rat heart were studied after 30 min of global ischemia. Functional recovery was assessed by the percentage recovery of developed pressure after 30 min reperfusion and by the magnitude of the contracture on reperfusion. After a control ischemia, developed pressure recovered to only 12+/-2% of pre-ischemic control and the reperfusion contracture was very large (81+/-6 mmHg). Activation of the mitochondrial KATP channel with 100 microM diazoxide present throughout ischemia and reperfusion improved recovery of developed pressure to 36+/-3% and reduced the reperfusion contracture (53+/-4 mmHg). Inhibition of the sodium/hydrogen exchanger with 10 microM cariporide caused a larger recovery of developed pressure to 72+/-4% and further reduced the reperfusion contracture (11+/-3 mmHg). The combination of both drugs increased recovery of developed pressure to 96+/-4% and the reperfusion contracture remained small (11+/-5 mmHg). The effectiveness of the timing of exposure to these drugs was explored. When both diazoxide and cariporide were applied 2 min before the end of ischaemia and remained present during reperfusion the recovery of developed pressure was 81+/-4% and the reperfusion contracture was small (12+/-3 mmHg); neither was significantly different to the recovery when both drugs were present throughout ischemia and reperfusion. We conclude that mitochondrial damage, blocked by diazoxide, and the coupled exchanger pathway, blocked by cariporide, are two of the principal damage pathways and functional recovery appears to be complete when both are blocked. The combination of these drugs is also highly effective when given 2 min before the end of ischemia.
Collapse
Affiliation(s)
- Xiao-Hui Xiao
- Department of Physiology and Institute for Biomedical Research, University of Sydney F13, NSW 2006, Australia
| | | |
Collapse
|
122
|
Masini E, Pierpaoli S, Marzocca C, Mannaioni PF, Pietrangeli P, Mateescu MA, Zelli M, Federico R, Mondovì B. Protective effects of a plant histaminase in myocardial ischaemia and reperfusion injury in vivo. Biochem Biophys Res Commun 2003; 309:432-9. [PMID: 12951068 DOI: 10.1016/j.bbrc.2003.08.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Grass pea seedling histaminase (a copper-diamine oxidase) was found to exert a significant cardioprotection against post-ischaemic reperfusion damage. Electrocardiogram (ECG) recordings from the rats subjected in vivo to ischaemia and reperfusion showed ventricular tachycardia (VT) and ventricular fibrillations (VF) occurring in 9 out of 12 untreated rats whereas no ventricular arrhythmias were found under histaminase (80U/kg body weight) treatment (n=16 rats). Computer-assisted morphometry of the ischaemic reperfused hearts stained with nitroblue tetrazolium showed the extension of damaged myocardium (area at risk and infarct size) significantly reduced in rats treated with histaminase, in comparison with the non-treated rats, whereas no protection was found with the semicarbazide inactivated histaminase. Biochemical markers of ischaemia-reperfusion myocardial tissue damage: malonyldialdehyde (MDA), tissue calcium concentration, myeloperoxidase (MPO), and apoptosis indicator caspase-3 were significantly elevated in untreated post-ischaemic reperfused rats, but significantly reduced under histaminase protection. In conclusion, plant histaminase appears to protect hearts from ischaemia-reperfusion injury by more than one mechanism, essentially involving histamine oxidation, and possibly as reactive oxygen species scavenger, presenting good perspectives for a novel therapeutic approach in treatment of ischaemic heart pathology.
Collapse
Affiliation(s)
- Emanuela Masini
- Department of Preclinical and Clinical Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
Mitochondria play a key role in determining cell fate during exposure to stress. Their role during ischemia/reperfusion is particularly critical because of the conditions that promote both apoptosis by the mitochondrial pathway and necrosis by irreversible damage to mitochondria in association with mitochondrial permeability transition (MPT). MPT is caused by the opening of permeability transition pores in the inner mitochondrial membrane, leading to matrix swelling, outer membrane rupture, release of apoptotic signaling molecules such as cytochrome c from the intermembrane space, and irreversible injury to the mitochondria. During ischemia (the MPT priming phase), factors such as intracellular Ca2+ accumulation, long-chain fatty acid accumulation, and reactive oxygen species progressively increase mitochondrial susceptibility to MPT, increasing the likelihood that MPT will occur on reperfusion (the MPT trigger phase). Because functional cardiac recovery ultimately depends on mitochondrial recovery, cardioprotection by ischemic and pharmacological preconditioning must ultimately involve the prevention of MPT. Investigations into this area are beginning to unravel some of the mechanistic links between cardioprotective signaling and mitochondria.
Collapse
Affiliation(s)
- James N Weiss
- Cardiovascular Research Laboratory, Department of Medicine (Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, Calif 90095-1760, USA.
| | | | | | | |
Collapse
|
124
|
Teshima Y, Akao M, Li RA, Chong TH, Baumgartner WA, Johnston MV, Marbán E. Mitochondrial ATP-sensitive potassium channel activation protects cerebellar granule neurons from apoptosis induced by oxidative stress. Stroke 2003; 34:1796-802. [PMID: 12791941 DOI: 10.1161/01.str.0000077017.60947.ae] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Mitochondrial ATP-sensitive potassium (mitoK(ATP)) channels are present in the brain, and several reports have shown that mitoK(ATP) channel openers protect the brain against ischemic injury. However, the precise mechanisms of this protection are not well established. We hypothesized that mitoK(ATP) channel openers prevent apoptosis by preserving mitochondrial membrane potential. METHODS We investigated the effect of mitoK(ATP) channel openers on apoptosis induced by oxidative stress using cultured cerebellar granule neurons. RESULTS The mitoK(ATP) channel opener diazoxide (100 micromol/L) significantly suppressed the number of cells with terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL)-positive nuclei and the increase in caspase-3 activity induced by 20 micromol/L H2O2. Diazoxide and another opener, pinacidil, prevented the loss of mitochondrial inner membrane potential (Delta(Psi)m) induced by H2O2. These effects were abolished by 5-hydroxydecanoate (500 micromol/L), a mitoK(ATP) channel blocker. Cyclosporin A and bongkrekic acid, inhibitors of the mitochondrial permeability transition pore, also prevented Delta(Psi)m loss, confirming the involvement of the mitochondrial permeability transition in the apoptotic cascade in neurons. Furthermore, diazoxide prevented the increase in extracellular glutamate concentration induced by H2O2, but this effect was not attributable to activation of surface K(ATP) channels. CONCLUSIONS MitoK(ATP) channel openers inhibited apoptosis by preserving mitochondrial inner membrane potential. These beneficial effects may suggest a possible new target for neuroprotection.
Collapse
Affiliation(s)
- Yasushi Teshima
- Institute of Molecular Cardiobiology, The Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|
125
|
Korge P, Honda HM, Weiss JN. Effects of fatty acids in isolated mitochondria: implications for ischemic injury and cardioprotection. Am J Physiol Heart Circ Physiol 2003; 285:H259-69. [PMID: 12793979 DOI: 10.1152/ajpheart.01028.2002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Fatty acids accumulate during myocardial ischemia and are implicated in ischemia-reperfusion injury and mitochondrial dysfunction. Because functional recovery after ischemia-reperfusion ultimately depends on the ability of the mitochondria to recover membrane potential (DeltaPsim), we studied the effects of fatty acids on DeltaPsim regulation, cytochrome c release, and Ca2+ handling in isolated mitochondria under conditions that mimicked aspects of ischemia-reperfusion. Long-chain but not short-chain free fatty acids caused a progressive and reversible (with BSA) increase in inner membrane leakiness (proton leak), which limited mitochondrial ability to support DeltaPsim. In comparison, long-chain activated fatty acids promoted 1). a slower depolarization that was not reversible with BSA, 2). cytochrome c loss that was unrelated to permeability transition pore opening, and 3). inhibition of the adenine nucleotide translocator. Together, these results impaired both mitochondrial ATP production and Ca2+ handling. Diazoxide, a selective opener of mitochondrial ATP-dependent potassium (KATP) channels, partially protected against these effects. These findings indicate that long-chain fatty acid accumulation during ischemia-reperfusion may predispose mitochondria to cytochrome c loss and irreversible injury and identify a novel cardioprotective action of diazoxide.
Collapse
Affiliation(s)
- Paavo Korge
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, California 90095-17690, USA.
| | | | | |
Collapse
|
126
|
Javadov SA, Clarke S, Das M, Griffiths EJ, Lim KHH, Halestrap AP. Ischaemic preconditioning inhibits opening of mitochondrial permeability transition pores in the reperfused rat heart. J Physiol 2003; 549:513-24. [PMID: 12692185 PMCID: PMC2342939 DOI: 10.1113/jphysiol.2003.034231] [Citation(s) in RCA: 252] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Opening of the mitochondrial permeability transition pore (MPTP) is thought to be a critical event in mediating the damage to hearts that accompanies their reperfusion following prolonged ischaemia. Protection from reperfusion injury occurs if the prolonged ischaemic period is preceded by short ischaemic periods followed by recovery. Here we investigate whether such ischaemic preconditioning (IPC) is accompanied by inhibition of MPTP opening. MPTP opening in Langendorff-perfused rat hearts was determined by perfusion with 2-deoxy[3H]glucose ([3H]DOG) and measurement of mitochondrial [3H]DOG entrapment. We demonstrate that IPC inhibits initial MPTP opening in hearts reperfused after 30 min global ischaemia, and subsequently enhances pore closure as hearts recover. However, MPTP opening in mitochondria isolated from IPC hearts occurred more readily than control mitochondria, implying that MPTP inhibition by IPC in situ was secondary to other factors such as decreased calcium overload and oxidative stress. Hearts perfused with cyclosporin A or sanglifehrin A, powerful inhibitors of the MPTP, also recovered better from ischaemia than controls (improved haemodynamic function and less lactate dehydrogenase release). However, the mitochondrial DOG entrapment technique showed these agents to be less effective than IPC at preventing MPTP opening. Our data suggest that protection from reperfusion injury is better achieved by reducing factors that induce MPTP opening than by inhibiting the MPTP directly.
Collapse
Affiliation(s)
- Sabzali A Javadov
- Department of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | | | | | |
Collapse
|
127
|
Ichinose M, Yonemochi H, Sato T, Saikawa T. Diazoxide triggers cardioprotection against apoptosis induced by oxidative stress. Am J Physiol Heart Circ Physiol 2003; 284:H2235-41. [PMID: 12623783 DOI: 10.1152/ajpheart.01073.2002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although mitochondrial ATP-sensitive potassium (mitoK(ATP)) channels have been reported to reduce the extent of apoptosis, the critical timing of mitoK(ATP) channel opening required to protect myocytes against apoptosis remains unclear. In the present study, we examined whether the mitoK(ATP) channel serves as a trigger of cardioprotection against apoptosis induced by oxidative stress. Apoptosis of cultured neonatal rat cardiomyocytes was determined by flow cytometry (light scatter and propidium iodide/annexin V-FITC fluorescence) and by nuclear staining with Hoechst 33342. Mitochondrial membrane potential (DeltaPsi) was measured by flow cytometry of cells stained with rhodamine-123 (Rh-123). Exposure to H(2)O(2) (500 microM) induced apoptosis, and the percentage of apoptotic cells increased progressively and peaked at 2 h. This H(2)O(2)-induced apoptosis was associated with the loss of DeltaPsi, and the time course of decrease in Rh-123 fluorescence paralleled that of apoptosis. Pretreatment of cardiomyocytes with diazoxide (100 microM), a putative mitoK(ATP) channel opener, for 30 min before exposure to H(2)O(2) elicited transient and mild depolarization of DeltaPsi and consequently suppressed both apoptosis and DeltaPsi loss after 2-h exposure to H(2)O(2). These protective effects of diazoxide were abrogated by the mitoK(ATP) channel blocker 5-hydroxydecanoate (500 microM) but not by the sarcolemmal K(ATP) channel blocker HMR-1098 (30 microM). Our results suggest for the first time that diazoxide-induced opening of mitoK(ATP) channels triggers cardioprotection against apoptosis induced by oxidative stress in rat cardiomyocytes.
Collapse
Affiliation(s)
- Masashi Ichinose
- Department of Laboratory Medicine, Oita Medical University, Japan
| | | | | | | |
Collapse
|
128
|
Hodgson DM, Zingman LV, Kane GC, Perez-Terzic C, Bienengraeber M, Ozcan C, Gumina RJ, Pucar D, O'Coclain F, Mann DL, Alekseev AE, Terzic A. Cellular remodeling in heart failure disrupts K(ATP) channel-dependent stress tolerance. EMBO J 2003; 22:1732-42. [PMID: 12682006 PMCID: PMC154482 DOI: 10.1093/emboj/cdg192] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
ATP-sensitive potassium (K(ATP)) channels are required for maintenance of homeostasis during the metabolically demanding adaptive response to stress. However, in disease, the effect of cellular remodeling on K(ATP) channel behavior and associated tolerance to metabolic insult is unknown. Here, transgenic expression of tumor necrosis factor alpha induced heart failure with typical cardiac structural and energetic alterations. In this paradigm of disease remodeling, K(ATP) channels responded aberrantly to metabolic signals despite intact intrinsic channel properties, implicating defects proximal to the channel. Indeed, cardiomyocytes from failing hearts exhibited mitochondrial and creatine kinase deficits, and thus a reduced potential for metabolic signal generation and transmission. Consequently, K(ATP) channels failed to properly translate cellular distress under metabolic challenge into a protective membrane response. Failing hearts were excessively vulnerable to metabolic insult, demonstrating cardiomyocyte calcium loading and myofibrillar contraction banding, with tolerance improved by K(ATP) channel openers. Thus, disease-induced K(ATP) channel metabolic dysregulation is a contributor to the pathobiology of heart failure, illustrating a mechanism for acquired channelopathy.
Collapse
Affiliation(s)
- Denice M Hodgson
- Department of Medicine, Mayo Clinic, Mayo Foundation, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Kopustinskiene DM, Toleikis A, Saris NEL. Adenine nucleotide translocase mediates the K(ATP)-channel-openers-induced proton and potassium flux to the mitochondrial matrix. J Bioenerg Biomembr 2003; 35:141-8. [PMID: 12887012 DOI: 10.1023/a:1023746103401] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
KATP channel openers have been shown to protect ischemic-reperfused myocardium by mimicking ischemic preconditioning, although their mechanisms of action have not been fully clarified. In this study we investigated the influence of the adenine nucleotide translocase (ANT) inhibitors--carboxyatractyloside (CAT) and bongkrekic acid (BA)--on the diazoxide- and pinacidil-induced uncoupling of isolated rat heart mitochondria respiring on pyruvate and malate (6 + 6 mM). We found that both CAT (1.3 microM) and BA (20 microM) markedly reduced the uncoupling of mitochondrial oxidative phosphorylation induced by the K(ATP) channel openers. Thus, the uncoupling effect of diazoxide and pinacidil is evident only when ANT is not fixed by inhibitors in neither the C- nor the M-conformation. Moreover, the uncoupling effect of diazoxide and pinacidil was diminished in the presence of ADP or ATP, indicating a competition of K(ATP) channel openers with adenine nucleotides. CAT also abolished K+-dependent mitochondrial respiratory changes. Thus ANT could also be involved in the regulation of K(ATP)-channel-openers-induced K+ flux through the inner mitochondrial membrane.
Collapse
Affiliation(s)
- Dalia M Kopustinskiene
- Institute for Biomedical Research, Kaunas University of Medicine, Eiveniu Street 4, LT-3007, Kaunas, Lithuania.
| | | | | |
Collapse
|
130
|
Dzeja PP, Bast P, Ozcan C, Valverde A, Holmuhamedov EL, Van Wylen DGL, Terzic A. Targeting nucleotide-requiring enzymes: implications for diazoxide-induced cardioprotection. Am J Physiol Heart Circ Physiol 2003; 284:H1048-56. [PMID: 12666660 DOI: 10.1152/ajpheart.00847.2002] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Modulation of mitochondrial respiratory chain, dehydrogenase, and nucleotide-metabolizing enzyme activities is fundamental to cellular protection. Here, we demonstrate that the potassium channel opener diazoxide, within its cardioprotective concentration range, modulated the activity of flavin adenine dinucleotide-dependent succinate dehydrogenase with an IC50 of 32 microM and reduced the rate of succinate-supported generation of reactive oxygen species (ROS) in heart mitochondria. 5-Hydroxydecanoic fatty acid circumvented diazoxide-inhibited succinate dehydrogenase-driven electron flow, indicating a metabolism-dependent supply of redox equivalents to the respiratory chain. In perfused rat hearts, diazoxide diminished the generation of malondialdehyde, a marker of oxidative stress, which, however, increased on diazoxide washout. This effect of diazoxide mimicked ischemic preconditioning and was associated with reduced oxidative damage on ischemia-reperfusion. Diazoxide reduced cellular and mitochondrial ATPase activities, along with nucleotide degradation, contributing to preservation of myocardial ATP levels during ischemia. Thus, by targeting nucleotide-requiring enzymes, particularly mitochondrial succinate dehydrogenase and cellular ATPases, diazoxide reduces ROS generation and nucleotide degradation, resulting in preservation of myocardial energetics under stress.
Collapse
Affiliation(s)
- Petras P Dzeja
- Department of Medicine, Mayo Clinic, Mayo Foundation, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
131
|
Akao M, O'Rourke B, Kusuoka H, Teshima Y, Jones SP, Marbán E. Differential actions of cardioprotective agents on the mitochondrial death pathway. Circ Res 2003; 92:195-202. [PMID: 12574147 DOI: 10.1161/01.res.0000051862.16691.f9] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined the effect of cardioprotective agents on three distinct phases of the H2O2-induced response that leads to loss of mitochondrial membrane potential (DeltaPsi(m)) and cell death in cultured cardiac myocytes: (1) priming, consisting of calcium-dependent morphological changes in mitochondria (swelling and loss of cristae), with preserved DeltaPsi(m), (2) depolarization, the rapid DeltaPsi(m) depolarization caused by mitochondrial permeability transition pore (PTP) opening, and (3) cell fragmentation. The mitochondrial ATP-sensitive potassium (mitoK(ATP)) channel opener diazoxide markedly decreased the likelihood that cells would undergo priming: many mitochondria remained fully polarized and morphologically intact. Diazoxide not only decreased the number of cells undergoing DeltaPsi(m) depolarization but also delayed the onset of DeltaPsi(m) loss, whereas it did not change the duration of depolarization in unprotected cells. The adenine nucleotide translocase inhibitor bongkrekic acid mimicked the effect of diazoxide to suppress priming, except that its effects were not blocked by the mitoK(ATP) channel blocker 5-hydroxydecanoate. In contrast, the PTP inhibitor cyclosporin A (CsA) did not prevent priming: neither latency for DeltaPsi(m) depolarization nor mitochondrial morphological changes were affected. However, CsA slowed the process of depolarization and blunted its severity. Importantly, coapplication of diazoxide and CsA exhibited additive effects, improving the efficacy of protection. Activation of mitoK(ATP) channels suppresses the cell death process at its earliest stage, by preserving mitochondrial integrity during oxidative stress. By virtue of its pharmacology and its phenotypic consequences, this mode of action is distinguishable from that of other cardioprotective interventions.
Collapse
Affiliation(s)
- Masaharu Akao
- Institute of Molecular Cardiobiology, The Johns Hopkins University, Baltimore, Md 21205, USA
| | | | | | | | | | | |
Collapse
|
132
|
Baumann P, Poitry S, Roatti A, Baertschi AJ. Plasmalemmal KATP channels shape triggered calcium transients in metabolically impaired rat atrial myocytes. Am J Physiol Heart Circ Physiol 2002; 283:H2296-305. [PMID: 12388309 DOI: 10.1152/ajpheart.00393.2002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The relative role of plasmalemmal and mitochondrial ATP-sensitive K(+) (K(ATP)) channels in calcium homeostasis of the atrium is little understood. Electrically triggered (1 Hz) cytoplasmic calcium transients were measured by 340-to-380-nm wavelength fura 2 emission ratios in cultured rat atrial myocytes. CCCP, a mitochondrial protonophore (100-400 nmol/l), dose dependently reduced the transient amplitude by up to 85%, caused a slow rise in baseline calcium, and reduced the recovery time constant of the transient from 143 to 91 ms (P < 0.05). However, neither 5-hydroxydecanoate, a mitochondrial K(ATP) channel blocker, nor diazoxide (500 micromol/l) affected the amplitude, baseline, or time constant in CCCP-treated cells. HMR-1098 (30 micromol/l), a plasmalemmal K(ATP) channel blocker, and glibenclamide (1 micromol/l) increased the amplitude in CCCP-treated myocytes by 69-82%, sharply elevated the calcium baseline, and prolonged the recovery time constant to 181-193 ms (P < 0.01). Thus opening of plasmalemmal but not mitochondrial K(ATP) channels reduces the calcium overload in metabolically compromised but otherwise intact atrial myocytes. Mitochondrial K(ATP) channels probably operate through a different mechanism to afford ischemic protection.
Collapse
Affiliation(s)
- Philippe Baumann
- Department of Physiology, Centre Médical Universitaire, 1211 Geneva 4, Switzerland
| | | | | | | |
Collapse
|
133
|
Samavati L, Monick MM, Sanlioglu S, Buettner GR, Oberley LW, Hunninghake GW. Mitochondrial K(ATP) channel openers activate the ERK kinase by an oxidant-dependent mechanism. Am J Physiol Cell Physiol 2002; 283:C273-81. [PMID: 12055096 DOI: 10.1152/ajpcell.00514.2001] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Extracellular signal-regulated kinases (ERKs) are key regulatory proteins that mediate cell survival, proliferation, and differentiation. Reactive oxygen species (ROS) may play a role in activation of the ERK pathway. Because mitochondria are a major source of ROS, we investigated whether mitochondria-derived ROS play a role in ERK activation. Diazoxide, a potent mitochondrial ATP-sensitive K+ (K(ATP)) channel opener, is known to depolarize the mitochondrial membrane potential and cause a reversible oxidation of respiratory chain flavoproteins, thus increasing mitochondrial ROS production. Using THP-1 cells as a model, we postulated that opening mitochondrial K(ATP) channels would increase production of ROS and, thereby, regulate the activity of the ERK kinase. We found that opening mitochondrial K(ATP) channels by diazoxide induced production of ROS as determined by an increased rate of dihydroethidium and dichlorofluorescein fluorescence. This increased production of ROS was associated with increased phosphorylation of ERK kinase in a time-dependent fashion. The MEK inhibitors PD-98059 and U-0126 blocked ERK activation mediated by diazoxide. N-acetylcysteine, but not diphenyleneiodonium, attenuated ERK activation mediated by diazoxide. Adenovirus-mediated overexpression of manganese superoxide dismutase, which is expressed in mitochondria, decreased the rate of dihydroethidium oxidation as well as ERK activation. We conclude that mitochondrial K(ATP) channel openers trigger ERK activation via mitochondria-derived ROS.
Collapse
Affiliation(s)
- Lobelia Samavati
- Department of Medicine, University of Iowa College of Medicine and Veterans Administration Medical Center, Iowa City, Iowa 52242, USA.
| | | | | | | | | | | |
Collapse
|
134
|
Korge P, Honda HM, Weiss JN. Protection of cardiac mitochondria by diazoxide and protein kinase C: implications for ischemic preconditioning. Proc Natl Acad Sci U S A 2002; 99:3312-7. [PMID: 11867760 PMCID: PMC122515 DOI: 10.1073/pnas.052713199] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mitochondrial ATP-sensitive K (mitoK(ATP)) channels play a central role in protecting the heart from injury in ischemic preconditioning. In isolated mitochondria exposed to elevated extramitochondrial Ca, P(i), and anoxia to simulate ischemic conditions, the selective mitoK(ATP) channel agonist diazoxide (25-50 microM) potently reduced mitochondrial injury by preventing both the mitochondrial permeability transition (MPT) and cytochrome c loss from the intermembrane space. Both effects were blocked completely by the selective mitoK(ATP) antagonist 5-hydroxydecanoate. The protective effect against Ca-induced MPT was most evident under conditions in which the ability of electron transport to support membrane potential (Deltapsi(m)) was decreased and inner membrane leakiness was increased moderately. Under these conditions, mitoK(ATP) channel activity strongly regulated Deltapsi(m), and diazoxide prevented MPT by inhibiting the driving force for Ca uptake. Phorbol 12-myristate 13-acetate mimicked the protective effects of diazoxide, unless 5-hydroxydecanoate was present, indicating that protein kinase C activation also protects mitochondria by activating mitoK(ATP) channels. Because Deltapsi(m) recovery ultimately is required for heart functional recovery, these results may explain how mitoK(ATP) channel activation mimics ischemic preconditioning by protecting mitochondria as they pass through a critical vulnerability window during ischemia/reperfusion.
Collapse
Affiliation(s)
- Paavo Korge
- University of California Cardiovascular Research Laboratory and Department of Medicine (Cardiology), University of California School of Medicine, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|