Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Basic Res Cardiol ; 118(1): 26, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37400630

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a major public health concern. Its outcome is poor and, as of today, barely any treatments have been able to decrease its morbidity or mortality. Cardiosphere-derived cells (CDCs) are heart cell products with anti-fibrotic, anti-inflammatory and angiogenic properties. Here, we tested the efficacy of CDCs in improving left ventricular (LV) structure and function in pigs with HFpEF. Fourteen chronically instrumented pigs received continuous angiotensin II infusion for 5 weeks. LV function was investigated through hemodynamic measurements and echocardiography at baseline, after 3 weeks of angiotensin II infusion before three-vessel intra-coronary CDC (n = 6) or placebo (n = 8) administration and 2 weeks after treatment (i.e., at completion of the protocol). As expected, arterial pressure was significantly and similarly increased in both groups. This was accompanied by LV hypertrophy that was not affected by CDCs. LV systolic function remained similarly preserved during the whole protocol in both groups. In contrast, LV diastolic function was impaired (increases in Tau, LV end-diastolic pressure as well as E/A, E/E'septal and E/E'lateral ratios) but CDC treatment significantly improved all of these parameters. The beneficial effect of CDCs on LV diastolic function was not explained by reduced LV hypertrophy or increased arteriolar density; however, interstitial fibrosis was markedly reduced. Three-vessel intra-coronary administration of CDCs improves LV diastolic function and reduces LV fibrosis in this hypertensive model of HFpEF.


Subject(s)
Heart Failure , Animals , Angiotensin II , Fibrosis , Hypertrophy, Left Ventricular , Stroke Volume , Swine , Ventricular Function, Left
3.
Fundam Clin Pharmacol ; 37(4): 739-752, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36797226

ABSTRACT

Mitochondrial permeability transition pore (mPTP) opening is a critical event leading to cell injury during myocardial ischemia-reperfusion but having a reliable cellular model to study the effect of drugs targeting mPTP is an unmet need. This study evaluated whether the Ca2+ electrogenic ionophore ferutinin is a relevant tool to induce mPTP in cardiomyocytes. mPTP opening was monitored using the calcein/cobalt fluorescence technique in adult cardiomyocytes isolated from wild-type and cyclophylin D (CypD) knock-out mice. Concomitantly, the effect of ferutinin was assessed in isolated myocardial mitochondria. Our results confirmed the Ca2+ ionophoric effect of ferutinin in isolated mitochondria and cardiomyocytes. Ferutinin induced all the hallmarks of mPTP opening in cells (loss of calcein, of mitochondrial potential and cell death), but none of them could be inhibited by CypD deletion or cyclosporine A, indicating that mPTP opening was not the major contributor to the effect of ferutinin. This was confirmed in isolated mitochondria where ferutinin acts by different mechanisms dependent and independent of the mitochondrial membrane potential. At low ferutinin/mitochondria concentration ratio, ferutinin displays protonophoric-like properties, lowering the mitochondrial membrane potential and limiting oxidative phosphorylation without mitochondrial swelling. At high ferutinin/mitochondria ratio, ferutinin induced a sudden Ca2+ independent mitochondrial swelling, which is only partially inhibited by cyclosporine A. Together, these result show that ferutinin is not a suitable tool to investigate CypD-dependent mPTP opening in isolated cardiomyocytes because it possesses other mitochondrial properties such as swelling induction and mitochondrial uncoupling properties which impede its utilization.


Subject(s)
Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Mice , Animals , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Permeability Transition Pore/pharmacology , Mitochondrial Membrane Transport Proteins/metabolism , Cyclosporine/pharmacology , Cyclosporine/metabolism , Myocytes, Cardiac , Mitochondria, Heart/metabolism , Mice, Knockout , Calcium/metabolism
4.
J Am Coll Cardiol ; 80(23): 2205-2219, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36456051

ABSTRACT

BACKGROUND: Patients with metabolic syndrome (MetS) have an increased risk of atrial fibrillation (AF). Impaired Ca2+ homeostasis and mitochondrial dysfunction have emerged as an arrhythmogenic substrate in both patients and animal models of MetS. Whether impaired mitochondrial Ca2+ handling underlies AF associated with MetS remains poorly explored. OBJECTIVES: The aim of this study was to determine the initial mechanisms related to AF susceptibility and mitochondrial dysfunction encountered in metabolic cardiomyopathy. METHODS: A total of 161 mice and 34 patients were studied. Mitochondrial Ca2+ and mitochondrial Ca2+ uniporter complex (MCUC) were investigated in right atrial tissue of patients with (n = 18) or without (n = 16) MetS and of C57Bl/6J mice fed with a high-fat sucrose diet (HFS) for 2 (n = 42) or 12 (n = 39) weeks. Susceptibility to AF was evaluated in isolated sinoatrial tissue and in vivo in mice. RESULTS: Increased expression of the MICUs subunits of the MCUC (1.00 ± 0.33 AU vs 1.29 ± 0.23 AU; P = 0.034) was associated with impaired mitochondrial Ca2+ uptake in patients (168.7 ± 31.3 nmol/min/mg vs 127.3 ± 18.4 nmol/min/mg; P = 0.026) and HFS mice (0.10 ± 0.04 ΔF/F0 × ms-1 vs 0.06 ± 0.03 ΔF/F0 × ms-1; P = 0.0086, and 0.15 ± 0.07 ΔF/F0 × ms-1 vs 0.046 ± 0.03 ΔF/F0 × ms-1; P = 0.0076 in 2- and 12-week HFS mice, respectively). HFS mice elicited a 70% increased susceptibility to AF. The MCUC agonist kaempferol restored MCUC activity in vitro and abolished the occurrence of AF in HFS mice. CONCLUSIONS: Impaired MCUC activity and mitochondrial Ca2+ homeostasis from the early stage of metabolic cardiomyopathy in mice lead to AF. Given that similar defects in cardiac mitochondrial Ca2+ handling are present in MetS patients, the modulation of the MCUC activity represents an attractive antiarrhythmic strategy.


Subject(s)
Atrial Appendage , Atrial Fibrillation , Metabolic Syndrome , Mice , Animals , Atrial Fibrillation/etiology , Calcium , Metabolic Syndrome/complications , Anti-Arrhythmia Agents , Mice, Inbred C57BL
5.
Shock ; 56(5): 857-864, 2021 11 01.
Article in English | MEDLINE | ID: mdl-33978607

ABSTRACT

ABSTRACT: Mitochondria is often considered as the common nexus of cardiac and cerebral dysfunction after cardiac arrest. Here, our goal was to determine whether the time course of cardiac and cerebral mitochondrial dysfunction is similar after shockable versus non-shockable cardiac arrest in rabbits. Anesthetized rabbits were submitted to 10 min of no-flow by ventricular fibrillation (VF group) or asphyxia (non-shockable group). They were euthanized at the end of the no-flow period or 30 min, 120 min, or 24 h after resuscitation for in vitro evaluation of oxygen consumption and calcium retention capacity. In the brain (cortex and hippocampus), moderate mitochondrial dysfunction was evidenced at the end of the no-flow period after both causes of cardiac arrest versus baseline. It partly recovered at 30 and 120 min after cardiac arrest, with lower calcium retention capacity and higher substrate-dependant oxygen consumption after VF versus non-shockable cardiac arrest. However, after 24 h of follow-up, mitochondrial dysfunction dramatically increased after both VF and non-shockable cardiac arrest, despite greater neurological dysfunction after the latter one. In the heart, mitochondrial dysfunction was also maximal after 24 h following resuscitation, with no significant difference among the causes of the cardiac arrest. During the earlier timing of evaluation, calcium retention capacity and ADP-dependant oxygen consumption were lower and higher, respectively, after non-shockable cardiac arrest versus VF. In conclusion, the kinetics of cardiac and cerebral mitochondrial dysfunction suggests that mitochondrial function does not play a major role in the early phase of the post-resuscitation process but is only involved in the longer pathophysiological events.


Subject(s)
Brain Diseases/physiopathology , Brain/ultrastructure , Heart Arrest/physiopathology , Mitochondria/physiology , Ventricular Fibrillation/physiopathology , Animals , Male , Mitochondria, Heart/physiology , Rabbits
6.
J Pharmacol Exp Ther ; 376(3): 348-357, 2021 03.
Article in English | MEDLINE | ID: mdl-33303698

ABSTRACT

Mitochondrial permeability transition pore (mPTP) opening is a key event in cell death during myocardial ischemia reperfusion. Inhibition of its modulator cyclophilin D (CypD) by cyclosporine A (CsA) reduces ischemia-reperfusion injury. The use of cyclosporine A in this indication is debated; however, targeting mPTP remains a major goal to achieve. We investigated the protective effects of a new original small-molecule cyclophilin inhibitor C31, which was specifically designed to target CypD. CypD peptidylprolyl cis-trans isomerase (PPIase) activity was assessed by the standard chemotrypsin-coupled assay. The effects of C31 on mPTP opening were investigated in isolated mouse cardiac mitochondria by measuring mitochondrial swelling and calcium retention capacity (CRC) in rat H9C2 cardiomyoblasts and in adult mouse cardiomyocytes by fluorescence microscopy in isolated perfused mouse hearts and ex vivo after drug infusion in mice. C31 potently inhibited CypD PPIase activity and mitochondrial swelling. C31 was more effective at increasing mitochondrial CRC than CsA and was still able to increase CRC in Ppif -/- (CypD-inactivated) cardiac mitochondria. C31 delayed both mPTP opening and cell death in cardiomyocytes subjected to hypoxia reoxygenation. However, high concentrations of both drugs were necessary to reduce mPTP opening in isolated perfused hearts, and neither CsA nor C31 inhibited mPTP opening in heart after in vivo infusion, underlying the importance of myocardial drug distribution for cardioprotection. C31 is an original inhibitor of mPTP opening involving both CypD-dependent and -independent mechanisms. It constitutes a promising new cytoprotective agent. Optimization of its pharmacokinetic properties is now required prior to its use against cardiac ischemia-reperfusion injury. SIGNIFICANCE STATEMENT: This study demonstrates that the new cyclophilin inhibitor C31 potently inhibits cardiac mitochondrial permeability transition pore (mPTP) opening in vitro and ex vivo. The dual mechanism of action of C31 allows the prevention of mPTP opening beyond cyclophilin D inhibition. Further development of the compound might bring promising drug candidates for cardioprotection. However, the lack of effect of both C31 and cyclosporine A after systemic administration demonstrates the difficulties of targeting myocardial mitochondria in vivo and should be taken into account in cardioprotective strategies.


Subject(s)
Heart/drug effects , Mitochondrial Permeability Transition Pore/antagonists & inhibitors , Myocardium/metabolism , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Animals , Biological Transport , Cytosol/drug effects , Cytosol/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardium/cytology , Pyrrolidines/metabolism
7.
Nanomedicine ; 30: 102295, 2020 11.
Article in English | MEDLINE | ID: mdl-32889047

ABSTRACT

Duchenne muscular dystrophy (DMD) is a debilitating disorder related to dystrophin encoding gene mutations, often associated with dilated cardiomyopathy. However, it is still unclear how dystrophin deficiency affects cardiac sarcomere remodeling and contractile dysfunction. We employed second harmonic generation (SHG) microscopy, a nonlinear optical imaging technique that allows studying contractile apparatus organization without histologic fixation and immunostaining. Images were acquired on alive DMD (mdx) and wild type cardiomyocytes at different ages and at various external calcium concentrations. An automated image processing was developed to identify individual myofibrils and extract data about their organization. We observed a structural aging-dependent remodeling in mdx cardiomyocytes affecting sarcomere sinuosity, orientation and length that could not be anticipated from standard optical imaging. These results revealed for the first time the interest of SHG to evaluate the intracellular and sarcomeric remodeling of DMD cardiac tissue in an age-dependent manner that could participate in progressive contractile dysfunction.


Subject(s)
Dystrophin/genetics , Myocytes, Cardiac/metabolism , Second Harmonic Generation Microscopy/methods , Animals , Cellular Senescence , Mice , Mice, Inbred C57BL , Muscular Dystrophy, Duchenne/genetics , Myocytes, Cardiac/pathology , Sarcomeres/pathology
8.
Front Cell Dev Biol ; 8: 609493, 2020.
Article in English | MEDLINE | ID: mdl-33569379

ABSTRACT

Besides skeletal muscle dysfunction, Duchenne muscular dystrophy (DMD) exhibits a progressive cardiomyopathy characterized by an impaired calcium (Ca2+) homeostasis and a mitochondrial dysfunction. Here we aimed to determine whether sarco-endoplasmic reticulum (SR/ER)-mitochondria interactions and mitochondrial function were impaired in dystrophic heart at the early stage of the pathology. For this purpose, ventricular cardiomyocytes and mitochondria were isolated from 3-month-old dystrophin-deficient mice (mdx mice). The number of contacts points between the SR/ER Ca2+ release channels (IP3R1) and the porine of the outer membrane of the mitochondria, VDAC1, measured using in situ proximity ligation assay, was greater in mdx cardiomyocytes. Expression levels of IP3R1 as well as the mitochondrial Ca2+ uniporter (MCU) and its regulated subunit, MICU1, were also increased in mdx heart. MICU2 expression was however unchanged. Furthermore, the mitochondrial Ca2+ uptake kinetics and the mitochondrial Ca2+ content were significantly increased. Meanwhile, the Ca2+-dependent pyruvate dehydrogenase phosphorylation was reduced, and its activity significantly increased. In Ca2+-free conditions, pyruvate-driven complex I respiration was decreased whereas in the presence of Ca2+, complex I-mediated respiration was boosted. Further, impaired complex I-mediated respiration was independent of its intrinsic activity or expression, which remains unchanged but is accompanied by an increase in mitochondrial reactive oxygen species production. Finally, mdx mice were treated with the complex I modulator metformin for 1 month. Metformin normalized the SR/ER-mitochondria interaction, decreased MICU1 expression and mitochondrial Ca2+ content, and enhanced complex I-driven respiration. In summary, before any sign of dilated cardiomyopathy, the DMD heart displays an aberrant SR/ER-mitochondria coupling with an increase mitochondrial Ca2+ homeostasis and a complex I dysfunction. Such remodeling could be reversed by metformin providing a novel therapeutic perspective in DMD.

9.
Gastroenterology ; 157(5): 1368-1382, 2019 11.
Article in English | MEDLINE | ID: mdl-31336123

ABSTRACT

BACKGROUND & AIMS: Hepatic ischemia/reperfusion injury is a complication of liver surgery that involves mitochondrial dysfunction resulting from mitochondrial permeability transition pore (mPTP) opening. Cyclophilin D (PPIF or CypD) is a peptidyl-prolyl cis-trans isomerase that regulates mPTP opening in the inner mitochondrial membrane. We investigated whether and how recently created small-molecule inhibitors of CypD prevent opening of the mPTP in hepatocytes and the resulting effects in cell models and livers of mice undergoing ischemia/reperfusion injury. METHODS: We measured the activity of 9 small-molecule inhibitors of cyclophilins in an assay of CypD activity. The effects of the small-molecule CypD inhibitors or vehicle on mPTP opening were assessed by measuring mitochondrial swelling and calcium retention in isolated liver mitochondria from C57BL/6J (wild-type) and Ppif-/- (CypD knockout) mice and in primary mouse and human hepatocytes by fluorescence microscopy. We induced ischemia/reperfusion injury in livers of mice given a small-molecule CypD inhibitor or vehicle before and during reperfusion and collected samples of blood and liver for histologic analysis. RESULTS: The compounds inhibited peptidyl-prolyl isomerase activity (half maximal inhibitory concentration values, 0.2-16.2 µmol/L) and, as a result, calcium-induced mitochondrial swelling, by preventing mPTP opening (half maximal inhibitory concentration values, 1.4-132 µmol/L) in a concentration-dependent manner. The most potent inhibitor (C31) bound CypD with high affinity and inhibited swelling in mitochondria from livers of wild-type and Ppif-/- mice (indicating an additional, CypD-independent effect on mPTP opening) and in primary human and mouse hepatocytes. Administration of C31 in mice with ischemia/reperfusion injury before and during reperfusion restored hepatic calcium retention capacity and oxidative phosphorylation parameters and reduced liver damage compared with vehicle. CONCLUSIONS: Recently created small-molecule inhibitors of CypD reduced calcium-induced swelling in mitochondria from mouse and human liver tissues. Administration of these compounds to mice during ischemia/reperfusion restored hepatic calcium retention capacity and oxidative phosphorylation parameters and reduced liver damage. These compounds might be developed to protect patients from ischemia/reperfusion injury after liver surgery or for other hepatic or nonhepatic disorders related to abnormal mPTP opening.


Subject(s)
Enzyme Inhibitors/pharmacology , Liver Diseases/prevention & control , Liver/drug effects , Mitochondria, Liver/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Peptidyl-Prolyl Isomerase F/antagonists & inhibitors , Reperfusion Injury/prevention & control , Animals , Calcium Signaling/drug effects , Cells, Cultured , Peptidyl-Prolyl Isomerase F/genetics , Peptidyl-Prolyl Isomerase F/metabolism , Cytoprotection , Disease Models, Animal , Humans , Liver/enzymology , Liver/pathology , Liver Diseases/enzymology , Liver Diseases/genetics , Liver Diseases/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/enzymology , Mitochondria, Liver/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction
10.
Free Radic Biol Med ; 137: 194-200, 2019 06.
Article in English | MEDLINE | ID: mdl-31047988

ABSTRACT

H11 kinase/Hsp22 (Hsp22) is a small heat shock protein, which, when overexpressed cardiac specifically in transgenic (TG) mice, induces stable left ventricular (LV) hypertrophy. Hsp22 also increases oxidative phosphorylation and mitochondrial reactive oxygen species (ROS) production, mechanisms mediating LV hypertrophy, senescence and reduced lifespan. Therefore, we investigated whether ROS production mediates LV hypertrophy, senescence and reduced life span in Hsp22 TG mice. Survival curves revealed that TG mice had a 48% reduction in their mean life span compared to wild type (WT) mice. This was associated with a significant increase in senescence markers, such as p16, p19 mRNA levels as well as the percentage of ß-galactosidase positive cells and telomerase activity. Oxidized (GSSG)/reduced (GSH) glutathione ratio, an indicator of oxidative stress, and ROS production from 3 major cellular sources was measured in cardiac tissue. Hearts from TG mice exhibited a decrease in GSH/GSSG ratio together with increased ROS production from all sources. To study the role of ROS, mice were treated with the antioxidant Tempol from weaning to their sacrifice. Chronic Tempol treatment abolished oxidative stress and overproduction of ROS, and reduced myocardial hypertrophy and Akt phosphorylation in TG mice. Tempol also significantly extended life span and prevented aging markers in TG mice. Taken together these results show that overexpression of Hsp22 increases oxidative stress responsible for the induction of hypertrophy and senescence and ultimately reduction in life span.


Subject(s)
Heat-Shock Proteins/metabolism , Hypertrophy, Left Ventricular/metabolism , Mitochondria/metabolism , Molecular Chaperones/metabolism , Myocardium/pathology , Animals , Antioxidants/administration & dosage , Cells, Cultured , Cellular Senescence , Cyclic N-Oxides/administration & dosage , Heat-Shock Proteins/genetics , Hypertrophy, Left Ventricular/genetics , Longevity , Male , Mice , Mice, Transgenic , Molecular Chaperones/genetics , Myocardium/metabolism , Oxidative Stress , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Spin Labels
11.
Aging Cell ; 17(4): e12793, 2018 08.
Article in English | MEDLINE | ID: mdl-29888494

ABSTRACT

The cellular mechanisms responsible for aging are poorly understood. Aging is considered as a degenerative process induced by the accumulation of cellular lesions leading progressively to organ dysfunction and death. The free radical theory of aging has long been considered the most relevant to explain the mechanisms of aging. As the mitochondrion is an important source of reactive oxygen species (ROS), this organelle is regarded as a key intracellular player in this process and a large amount of data supports the role of mitochondrial ROS production during aging. Thus, mitochondrial ROS, oxidative damage, aging, and aging-dependent diseases are strongly connected. However, other features of mitochondrial physiology and dysfunction have been recently implicated in the development of the aging process. Here, we examine the potential role of the mitochondrial permeability transition pore (mPTP) in normal aging and in aging-associated diseases.


Subject(s)
Cellular Senescence , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Humans , Mitochondrial Permeability Transition Pore , Reactive Oxygen Species/metabolism
12.
Biochem Pharmacol ; 142: 87-95, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28645478

ABSTRACT

A major cause of cell death during myocardial ischemia-reperfusion is mitochondrial dysfunction. We previously showed that the reperfusion of an ischemic myocardium was associated with an accumulation of cholesterol into mitochondria and a concomitant strong generation of auto-oxidized oxysterols. The inhibition of mitochondrial accumulation of cholesterol abolished the formation of oxysterols and prevented mitochondrial injury at reperfusion. The aim of this study was to investigate the impact of hypercholesterolemia on sterol and oxysterol accumulation in rat cardiac cytosols and mitochondria and to analyse the effect of the translocator protein ligand 4'-chlorodiazepam on this accumulation and mitochondrial function. Hypercholesterolemic ZDF fa/fa rats or normocholesterolemic lean rats were submitted to 30min of coronary artery occlusion followed by 15min reperfusion where cardiac cytosols and mitochondria were isolated. Hypercholesterolemia increased the cellular cardiac concentrations of cholesterol, cholesterol precursors and oxysterols both in cytosol and mitochondria in non-ischemic conditions. It also amplified the accumulation of all these compounds in cardiac cells and the alteration of mitochondrial function with ischemia-reperfusion. Administration of 4'-chlorodiazepam to ZDF fa/fa rats had no effect on the enhancement of sterols and oxysterols observed in the cytosols but inhibited cholesterol transfer to the mitochondria. It also alleviated the mitochondrial accumulation of all the investigated sterols and oxysterols. This was associated with a restoration of oxidative phosphorylation and a prevention of mitochondrial transition pore opening. The inhibition of cholesterol accumulation with TSPO ligands represents an interesting strategy to protect the mitochondria during ischemia-reperfusion in hypercholesterolemic conditions.


Subject(s)
Benzodiazepinones/pharmacology , Carrier Proteins/metabolism , Hypercholesterolemia/metabolism , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/prevention & control , Receptors, GABA-A/metabolism , Sterols/metabolism , Animals , Benzodiazepinones/therapeutic use , Cytosol/metabolism , Hypercholesterolemia/complications , Ligands , Male , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Oxidative Phosphorylation , Rats, Zucker
13.
Sci Rep ; 7(1): 4283, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28655872

ABSTRACT

Opening of the mitochondrial permeability transition pore (mPTP) plays a major role in cell death during cardiac ischaemia-reperfusion. Adult isolated rodent cardiomyocytes are valuable cells to study the effect of drugs targeting mPTP. This study investigated whether the use of Ca2+ ionophores (A23187, ionomycin and ETH129) represent a reliable model to study inhibition of mPTP opening in cardiomyocytes. We monitored mPTP opening using the calcein/cobalt fluorescence technique in adult rat and wild type or cyclophilin D (CypD) knock-out mice cardiomyocytes. Cells were either treated with Ca2+ ionophores or subjected to hypoxia followed by reoxygenation. The ionophores induced mPTP-dependent swelling in isolated mitochondria. A23187, but not ionomycin, induced a decrease in calcein fluorescence. This loss could not be inhibited by CypD deletion and was explained by a direct interaction between A23187 and cobalt. ETH129 caused calcein loss, mitochondrial depolarization and cell death but CypD deletion did not alleviate these effects. In the hypoxia-reoxygenation model, CypD deletion delayed both mPTP opening and cell death occurring at the time of reoxygenation. Thus, Ca2+ ionophores are not suitable to induce CypD-dependent mPTP opening in adult murine cardiomyocytes. Hypoxia-reoxygenation conditions appear therefore as the most reliable model to investigate mPTP opening in these cells.


Subject(s)
Calcium/metabolism , Ion Channel Gating/drug effects , Ionophores/pharmacology , Mitochondrial Membrane Transport Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Hypoxia/metabolism , Mice , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Permeability Transition Pore , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...