Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Circ Res ; 132(11): e171-e187, 2023 05 26.
Article in English | MEDLINE | ID: mdl-37057625

ABSTRACT

BACKGROUND: Cardiac contractile function requires high energy from mitochondria, and Ca2+ from the sarcoplasmic reticulum (SR). Via local Ca2+ transfer at close mitochondria-SR contacts, cardiac excitation feedforward regulates mitochondrial ATP production to match surges in demand (excitation-bioenergetics coupling). However, pathological stresses may cause mitochondrial Ca2+ overload, excessive reactive oxygen species production and permeability transition, risking homeostatic collapse and myocyte loss. Excitation-bioenergetics coupling involves mitochondria-SR tethers but the role of tethering in cardiac physiology/pathology is debated. Endogenous tether proteins are multifunctional; therefore, nonselective targets to scrutinize interorganelle linkage. Here, we assessed the physiological/pathological relevance of selective chronic enhancement of cardiac mitochondria-SR tethering. METHODS: We introduced to mice a cardiac muscle-specific engineered tether (linker) transgene with a fluorescent protein core and deployed 2D/3D electron microscopy, biochemical approaches, fluorescence imaging, in vivo and ex vivo cardiac performance monitoring and stress challenges to characterize the linker phenotype. RESULTS: Expressed in the mature cardiomyocytes, the linker expanded and tightened individual mitochondria-junctional SR contacts; but also evoked a marked remodeling with large dense mitochondrial clusters that excluded dyads. Yet, excitation-bioenergetics coupling remained well-preserved, likely due to more longitudinal mitochondria-dyad contacts and nanotunnelling between mitochondria exposed to junctional SR and those sealed away from junctional SR. Remarkably, the linker decreased female vulnerability to acute massive ß-adrenergic stress. It also reduced myocyte death and mitochondrial calcium-overload-associated myocardial impairment in ex vivo ischemia/reperfusion injury. CONCLUSIONS: We propose that mitochondria-SR/endoplasmic reticulum contacts operate at a structural optimum. Although acute changes in tethering may cause dysfunction, upon chronic enhancement of contacts from early life, adaptive remodeling of the organelles shifts the system to a new, stable structural optimum. This remodeling balances the individually enhanced mitochondrion-junctional SR crosstalk and excitation-bioenergetics coupling, by increasing the connected mitochondrial pool and, presumably, Ca2+/reactive oxygen species capacity, which then improves the resilience to stresses associated with dysregulated hyperactive Ca2+ signaling.


Subject(s)
Calcium Signaling , Sarcoplasmic Reticulum , Female , Mice , Animals , Sarcoplasmic Reticulum/metabolism , Reactive Oxygen Species/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Mitochondria, Heart/metabolism , Calcium/metabolism
2.
Aging Cell ; 21(3): e13564, 2022 03.
Article in English | MEDLINE | ID: mdl-35233924

ABSTRACT

Aged cardiomyocytes develop a mismatch between energy demand and supply, the severity of which determines the onset of heart failure, and become prone to undergo cell death. The FoF1-ATP synthase is the molecular machine that provides >90% of the ATP consumed by healthy cardiomyocytes and is proposed to form the mitochondrial permeability transition pore (mPTP), an energy-dissipating channel involved in cell death. We investigated whether aging alters FoF1-ATP synthase self-assembly, a fundamental biological process involved in mitochondrial cristae morphology and energy efficiency, and the functional consequences this may have. Purified heart mitochondria and cardiomyocytes from aging mice displayed an impaired dimerization of FoF1-ATP synthase (blue native and proximity ligation assay), associated with abnormal mitochondrial cristae tip curvature (TEM). Defective dimerization did not modify the in vitro hydrolase activity of FoF1-ATP synthase but reduced the efficiency of oxidative phosphorylation in intact mitochondria (in which membrane architecture plays a fundamental role) and increased cardiomyocytes' susceptibility to undergo energy collapse by mPTP. High throughput proteomics and fluorescence immunolabeling identified glycation of 5 subunits of FoF1-ATP synthase as the causative mechanism of the altered dimerization. In vitro induction of FoF1-ATP synthase glycation in H9c2 myoblasts recapitulated the age-related defective FoF1-ATP synthase assembly, reduced the relative contribution of oxidative phosphorylation to cell energy metabolism, and increased mPTP susceptibility. These results identify altered dimerization of FoF1-ATP synthase secondary to enzyme glycation as a novel pathophysiological mechanism involved in mitochondrial cristae remodeling, energy deficiency, and increased vulnerability of cardiomyocytes to undergo mitochondrial failure during aging.


Subject(s)
Aging , Mitochondria, Heart , Mitochondrial Proton-Translocating ATPases , Myocytes, Cardiac , Adenosine Triphosphate/metabolism , Aging/metabolism , Aging/physiology , Animals , Calcium/metabolism , Dimerization , Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/metabolism , Mice , Mitochondria, Heart/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/metabolism , Myocytes, Cardiac/metabolism
3.
Adv Sci (Weinh) ; 9(4): e2103265, 2022 02.
Article in English | MEDLINE | ID: mdl-34904402

ABSTRACT

Stroke is the second leading cause of death and disability worldwide. Current treatments, such as pharmacological thrombolysis or mechanical thrombectomy, reopen occluded arteries but do not protect against ischemia-induced damage that occurs before reperfusion or neuronal damage induced by ischemia/reperfusion. It has been shown that disrupting the conversion of glyoxal to glycolic acid (GA) results in a decreased tolerance to anhydrobiosis in Caenorhabditis elegans dauer larva and that GA itself can rescue this phenotype. During the process of desiccation/rehydration, a metabolic stop/start similar to the one observed during ischemia/reperfusion occurs. In this study, the protective effect of GA is tested in different ischemia models, i.e., in commonly used stroke models in mice and swine. The results show that GA, given during reperfusion, strongly protects against ischemic damage and improves functional outcome. Evidence that GA exerts its effect by counteracting the glutamate-dependent increase in intracellular calcium during excitotoxicity is provided. These results suggest that GA treatment has the potential to reduce mortality and disability in stroke patients.


Subject(s)
Brain Ischemia/drug therapy , Calcium/metabolism , Glycolates/pharmacology , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Animals , Brain Ischemia/metabolism , Desiccation , Disease Models, Animal , Glycolates/administration & dosage , Glycolates/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotection/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Reperfusion Injury/metabolism , Swine
4.
J Physiol ; 598(7): 1307-1326, 2020 04.
Article in English | MEDLINE | ID: mdl-30218574

ABSTRACT

Current dogma holds that the heart balances energy demand and supply effectively and sustainably by sequestering enough Ca2+ into mitochondria during heartbeats to stimulate metabolic enzymes in the tricarboxylic acid (TCA) cycle and electron transport chain (ETC). This process is called excitation-contraction-bioenergetics (ECB) coupling. Recent breakthroughs in identifying the mitochondrial Ca2+ uniporter (MCU) and its associated proteins have opened up new windows for interrogating the molecular mechanisms of mitochondrial Ca2+ homeostasis regulation and its role in ECB coupling. Despite remarkable progress made in the past 7 years, it has been surprising, almost disappointing, that germline MCU deficiency in mice with certain genetic background yields viable pups, and knockout of the MCU in adult heart does not cause lethality. Moreover, MCU deficiency results in few adverse phenotypes, normal performance, and preserved bioenergetics in the heart at baseline. In this review, we briefly assess the existing literature on mitochondrial Ca2+ homeostasis regulation and then we consider possible explanations for why MCU-deficient mice are spared from energy crises under physiological conditions. We propose that MCU and/or mitochondrial Ca2+ may have limited ability to set ECB coupling, that other mitochondrial Ca2+ handling mechanisms may play a role, and that extra-mitochondrial Ca2+ may regulate ECB coupling. Since the heart needs to regenerate a significant amount of ATP to assure the perpetuation of heartbeats, multiple mechanisms are likely to work in concert to match energy supply with demand.


Subject(s)
Calcium , Mitochondria , Animals , Calcium/metabolism , Energy Metabolism , Heart , Mice , Mitochondria/metabolism , Oxidation-Reduction
5.
FEBS Lett ; 593(13): 1528-1541, 2019 07.
Article in English | MEDLINE | ID: mdl-31058316

ABSTRACT

Intracellular Ca2+ signaling controls numerous cellular functions. Mitochondria respond to cytosolic Ca2+ changes by adapting mitochondrial functions and, in some cell types, shaping the spatiotemporal properties of the cytosolic Ca2+ signal. Numerous methods have been developed to specifically and quantitatively measure the mitochondrial-free Ca2+ concentrations ([Ca2+ ]m ), but there are still significant discrepancies in the calculated absolute values of [Ca2+ ]m in stimulated live cells. These discrepancies may be due to the distinct properties of the methods used to measure [Ca2+ ]m , the calcium-free/bound ratio, and the cell-type and stimulus-dependent Ca2+ dynamics. Critical processes happening in the mitochondria, such as ATP generation, ROS homeostasis, and mitochondrial permeability transition opening, depend directly on the [Ca2+ ]m values. Thus, precise determination of absolute [Ca2+ ]m values is imperative for understanding Ca2+ signaling. This review summarizes the reported calibrated [Ca2+ ]m values in many cell types and discusses the discrepancies among these values. Areas for future research are also proposed.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Animals , Cell Survival , Energy Metabolism , Humans
6.
Cell Rep ; 24(12): 3099-3107.e4, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30231993

ABSTRACT

Mitochondrial Ca2+ elevations enhance ATP production, but uptake must be balanced by efflux to avoid overload. Uptake is mediated by the mitochondrial Ca2+ uniporter channel complex (MCUC), and extrusion is controlled largely by the Na+/Ca2+ exchanger (NCLX), both driven electrogenically by the inner membrane potential (ΔΨm). MCUC forms hotspots at the cardiac mitochondria-junctional SR (jSR) association to locally receive Ca2+ signals; however, the distribution of NCLX is unknown. Our fractionation-based assays reveal that extensively jSR-associated mitochondrial segments contain a minor portion of NCLX and lack Na+-dependent Ca2+ extrusion. This pattern is retained upon in vivo NCLX overexpression, suggesting extensive targeting to non-jSR-associated submitochondrial domains and functional relevance. In cells with non-polarized MCUC distribution, upon NCLX overexpression the same given increase in matrix Ca2+ expends more ΔΨm. Thus, cardiac mitochondrial Ca2+ uptake and extrusion are reciprocally polarized, likely to optimize the energy efficiency of local calcium signaling in the beating heart.


Subject(s)
Calcium Signaling , Calcium/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Animals , Cell Line , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Sodium/metabolism
7.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt B): 1991-2001, 2018 May.
Article in English | MEDLINE | ID: mdl-28918113

ABSTRACT

Recent advancement in mitochondrial research has significantly extended our knowledge on the role and regulation of mitochondria in health and disease. One important breakthrough is the delineation of how mitochondrial morphological changes, termed mitochondrial dynamics, are coupled to the bioenergetics and signaling functions of mitochondria. In general, it is believed that fusion leads to an increased mitochondrial respiration efficiency and resistance to stress-induced dysfunction while fission does the contrary. This concept seems not applicable to adult cardiomyocytes. The mitochondria in adult cardiomyocytes exhibit fragmented morphology (tilted towards fission) and show less networking and movement as compared to other cell types. However, being the most energy-demanding cells, cardiomyocytes in the adult heart possess vast number of mitochondria, high level of energy flow, and abundant mitochondrial dynamics proteins. This apparent discrepancy could be explained by recently identified new functions of the mitochondrial dynamics proteins. These "non-canonical" roles of mitochondrial dynamics proteins range from controlling inter-organelle communication to regulating cell viability and survival under metabolic stresses. Here, we summarize the newly identified non-canonical roles of mitochondrial dynamics proteins. We focus on how these fission and fusion independent roles of dynamics proteins regulate mitochondrial bioenergetics. We also discuss potential molecular mechanisms, unique intracellular location, and the cardiovascular disease relevance of these non-canonical roles of the dynamics proteins. We propose that future studies are warranted to differentiate the canonical and non-canonical roles of dynamics proteins and to identify new approaches for the treatment of heart diseases. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.


Subject(s)
Energy Metabolism , Heart Diseases/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Signal Transduction
8.
J Biol Chem ; 291(44): 23343-23362, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27637331

ABSTRACT

Control of myocardial energetics by Ca2+ signal propagation to the mitochondrial matrix includes local Ca2+ delivery from sarcoplasmic reticulum (SR) ryanodine receptors (RyR2) to the inner mitochondrial membrane (IMM) Ca2+ uniporter (mtCU). mtCU activity in cardiac mitochondria is relatively low, whereas the IMM surface is large, due to extensive cristae folding. Hence, stochastically distributed mtCU may not suffice to support local Ca2+ transfer. We hypothesized that mtCU concentrated at mitochondria-SR associations would promote the effective Ca2+ transfer. mtCU distribution was determined by tracking MCU and EMRE, the proteins essential for channel formation. Both proteins were enriched in the IMM-outer mitochondrial membrane (OMM) contact point submitochondrial fraction and, as super-resolution microscopy revealed, located more to the mitochondrial periphery (inner boundary membrane) than inside the cristae, indicating high accessibility to cytosol-derived Ca2+ inputs. Furthermore, MCU immunofluorescence distribution was biased toward the mitochondria-SR interface (RyR2), and this bias was promoted by Ca2+ signaling activity in intact cardiomyocytes. The SR fraction of heart homogenate contains mitochondria with extensive SR associations, and these mitochondria are highly enriched in EMRE. Size exclusion chromatography suggested for EMRE- and MCU-containing complexes a wide size range and also revealed MCU-containing complexes devoid of EMRE (thus disabled) in the mitochondrial but not the SR fraction. Functional measurements suggested more effective mtCU-mediated Ca2+ uptake activity by the mitochondria of the SR than of the mitochondrial fraction. Thus, mtCU "hot spots" can be formed at the cardiac muscle mitochondria-SR associations via localization and assembly bias, serving local Ca2+ signaling and the excitation-energetics coupling.


Subject(s)
Calcium Channels/metabolism , Myocardium/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Signaling , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism
9.
J Biol Chem ; 291(3): 1221-34, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26582200

ABSTRACT

Death receptors are members of the tumor necrosis factor receptor superfamily involved in the extrinsic apoptotic pathway. Lifeguard (LFG) is a death receptor antagonist mainly expressed in the nervous system that specifically blocks Fas ligand (FasL)-induced apoptosis. To investigate its mechanism of action, we studied its subcellular localization and its interaction with members of the Bcl-2 family proteins. We performed an analysis of LFG subcellular localization in murine cortical neurons and found that LFG localizes mainly to the ER and Golgi. We confirmed these results with subcellular fractionation experiments. Moreover, we show by co-immunoprecipitation experiments that LFG interacts with Bcl-XL and Bcl-2, but not with Bax or Bak, and this interaction likely occurs in the endoplasmic reticulum. We further investigated the relationship between LFG and Bcl-XL in the inhibition of apoptosis and found that LFG protects only type II apoptotic cells from FasL-induced death in a Bcl-XL dependent manner. The observation that LFG itself is not located in mitochondria raises the question as to whether LFG in the ER participates in FasL-induced death. Indeed, we investigated the degree of calcium mobilization after FasL stimulation and found that LFG inhibits calcium release from the ER, a process that correlates with LFG blockage of cytochrome c release to the cytosol and caspase activation. On the basis of our observations, we propose that there is a required step in the induction of type II apoptotic cell death that involves calcium mobilization from the ER and that this step is modulated by LFG.


Subject(s)
Apoptosis , Calcium Signaling , Endoplasmic Reticulum/metabolism , Fas Ligand Protein/antagonists & inhibitors , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice, Inbred C57BL , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
10.
Exp Physiol ; 100(1): 23-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25557728

ABSTRACT

NEW FINDINGS: What is the central question of this study? The microtubule network is disrupted during myocardial ischaemia-reperfusion injury. It was suggested that prevention of microtubule disruption with paclitaxel might reduce cardiac infarct size; however, the effects on infarction have not been studied. What is the main finding and its importance? Paclitaxel caused a reduction in microtubule disruption and cardiomyocyte hypercontracture during ischaemia-reperfusion. However, it induced a greater increase in cytosolic calcium, which may explain the lack of effect against infarction that we have seen in isolated rat hearts. The large increase in perfusion pressure induced by paclitaxel in this model may have clinical implications, because detrimental effects of the drug were reported after its clinical application. Microtubules play a major role in the transmission of mechanical forces within the myocardium and in maintenance of organelle function. However, this intracellular network is disrupted during myocardial ischaemia-reperfusion. We assessed the effects of prevention of microtubule disruption with paclitaxel on ischaemia-reperfusion injury in isolated rat cardiomyocytes and hearts. Isolated rat cardiomyocytes were submitted to normoxia (1 h) or 45 min of simulated ischaemia (pH 6.4, 0% O2 , 37 °C) and reoxygenation, without or with treatment with the microtubule stabilizer, paclitaxel (10(-5) M), or the inhibitor of microtubule polymerization, colchicine (5 × 10(-6) M). Simulated ischaemia leads to microtubule disruption before the onset of ischaemic contracture. Paclitaxel attenuated both microtubule disruption and the incidence of hypercontracture, whereas treatment with colchicine mimicked the effects of simulated ischaemia and reoxygenation. In isolated normoxic rat hearts, treatment with paclitaxel induced concentration-dependent decreases in heart rate and left ventricular developed pressure and increases in perfusion pressure. Despite protection against hypercontracture, paclitaxel pretreatment did not modify infarct size (60.37 ± 2.27% in control hearts versus 58.75 ± 10.25, 55.44 ± 10.32 and 50.06 ± 10.14% after treatment with 10(-6) , 3 × 10(-6) and 10(-5) m of paclitaxel) after 60 min of global ischaemia and reperfusion in isolated rat hearts. Lack of protection was correlated with a higher increase in cytosolic calcium levels during simulated ischaemia in cardiomyocytes treated with paclitaxel (2.32 ± 0.15 versus 1.13 ± 0.16 a.u. in the presence or absence of 10(-6) m paclitaxel, respectively, P < 0.05), but not with changes in aortic reactivity. In conclusion, microtubule stabilization with paclitaxel reduces hypercontracture in isolated rat cardiomyocytes but does not protect against infarction in isolated rat hearts.


Subject(s)
Microtubules/drug effects , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Paclitaxel/pharmacology , Tubulin Modulators/pharmacology , Animals , Calcium Signaling/drug effects , Colchicine/pharmacology , Cytoprotection , Dose-Response Relationship, Drug , Heart Rate/drug effects , In Vitro Techniques , Male , Mechanotransduction, Cellular/drug effects , Microtubules/metabolism , Microtubules/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Stress, Mechanical , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
11.
Thromb Haemost ; 113(3): 441-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25631625

ABSTRACT

Aging is a major determinant of the incidence and severity of ischaemic heart disease. Preclinical information suggests the existence of intrinsic cellular alterations that contribute to ischaemic susceptibility in senescent myocardium, by mechanisms not well established. We investigated the role of altered mitochondrial function in the adverse effect of aging. Isolated perfused hearts from old mice (> 20 months) displayed increased ischaemia-reperfusion injury as compared to hearts from adult mice (6 months) despite delayed onset of ischaemic rigor contracture. In cardiomyocytes from aging hearts there was a more rapid decline of mitochondrial membrane potential (Δψm) as compared to young ones, but ischaemic rigor shortening was also delayed. Transient recovery of Δψm observed during ischaemia, secondary to the reversal of mitochondrial FoF1 ATP synthase to ATPase mode, was markedly reduced in aging cardiomyocytes. Proteomic analysis demonstrated increased oxidation of different subunits of ATP synthase. Altered bionergetics in aging cells was associated with reduced mitochondrial calcium uptake and more severe cytosolic calcium overload during ischaemia-reperfusion. Despite attenuated ROS burst and mitochondrial calcium overload, mitochondrial permeability transition pore (mPTP) opening and cell death was increased in reperfused aged cells. In vitro studies demonstrated a significantly reduced calcium retention capacity in interfibrillar mitochondria from aging hearts. Our results identify altered FoF1 ATP synthase and increased sensitivity of mitochondria to undergo mPTP opening as important determinants of the reduced tolerance to ischaemia-reperfusion in aging hearts. Because ATP synthase has been proposed to conform mPTP, it is tempting to hypothesise that oxidation of ATP synthase underlie both phenomena.


Subject(s)
Mitochondria, Heart/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Proton-Translocating ATPases/metabolism , Age Factors , Animals , Calcium/metabolism , Cell Death , Energy Metabolism , Isolated Heart Preparation , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mitochondria, Heart/pathology , Mitochondrial Permeability Transition Pore , Myocardial Contraction , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/pathology , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Sarcolemma/metabolism , Time Factors
12.
J Mol Cell Cardiol ; 68: 79-88, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24434643

ABSTRACT

Mitochondria play a central role in the protection conferred by ischemic preconditioning (IP) by not fully elucidated mechanisms. We investigated whether IP protects mitochondria against ischemia-reperfusion (IR) injury through mechanisms independent of cytosolic signaling. In isolated rat hearts, sublethal IR increased superoxide production and reduced complex-I- and II-mediated respiration in subsarcolemmal (SS), but not interfibrillar (IF) mitochondria. This effect of IR on mitochondrial respiration was significantly attenuated by IP. Similar results were obtained in isolated cardiac mitochondria subjected to in vitro IR. The reduction in SS mitochondrial respiration in the heart and in vitro model was paralleled by an increase in oxidized cysteine residues, which was also prevented by IP. IP was also protective in mitochondria submitted to lethal IR. The protective effect of IP against respiratory failure was unaffected by inhibition of mitochondrial KATP channels or mitochondrial permeability transition. However, IP protection was lost in mitochondria from genetically-modified animals in which connexin-43, a protein present in SS but not IF mitochondria, was replaced by connexin-32. Our results demonstrate the existence of a protective mitochondrial mechanism or "mitochondrial preconditioning" independent of cytosol that confers protection against IR-induced respiratory failure and oxidative damage, and requires connexin-43.


Subject(s)
Ischemic Preconditioning , Mitochondria, Heart/physiology , Myocardial Reperfusion Injury/prevention & control , Animals , Cell Respiration , Connexin 43/metabolism , Coronary Circulation , Cytosol/metabolism , In Vitro Techniques , Ion Channel Gating , Male , Mice , Mice, Knockout , Mitochondrial Proteins/metabolism , Myocardial Reperfusion Injury/pathology , Oxidation-Reduction , Oxidative Phosphorylation , Oxidative Stress , Peroxynitrous Acid/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Superoxides/metabolism
13.
Basic Res Cardiol ; 108(3): 351, 2013 May.
Article in English | MEDLINE | ID: mdl-23595215

ABSTRACT

Connexin 43 (Cx43) deficiency increases myocardial tolerance to ischemia-reperfusion injury and abolishes preconditioning protection. It is not known whether modifications in baseline signaling through protective RISK or SAFE pathways or in response to preconditioning may contribute to these effects. To answer this question we used Cx43(Cre-ER(T)/fl) mice, in which Cx43 expression is abolished after 4-hydroxytamoxifen (4-OHT) administration. Isolated hearts from Cx43(Cre-ER(T)/fl) mice, or from Cx43(fl/fl) controls, treated with vehicle or 4-OHT, were submitted to global ischemia (40 min) and reperfusion. Cx43 deficiency was associated with reduced infarct size after ischemia-reperfusion (11.17 ± 3.25 % vs. 65.04 ± 3.79, 59.31 ± 5.36 and 65.40 ± 4.91, in Cx43(fl/fl) animals treated with vehicle, Cx43(fl/fl) mice treated with 4-OHT, and Cx43(Cre-ER(T)/fl) mice treated with vehicle, respectively, n = 8-9, p < 0.001). However, the ratio phosphorylated/total protein expression for Akt, ERK-1/2, GSK3ß and STAT3 was not increased in normoxic samples from animals lacking Cx43. Instead, a reduction in the phosphorylation state of GSK3ß was observed in Cx43-deficient mice (ratio: 0.15 ± 0.02 vs. 0.56 ± 0.11, 0.77 ± 0.15, and 0.46 ± 0.14, respectively, n = 5-6, p < 0.01). Furthermore, ischemic preconditioning (IPC, 4 cycles of 3.5 min of ischemia and 5 min of reperfusion) increased phosphorylation of ERK-1/2, GSK3ß, and STAT3 in all hearts without differences between groups (n = 5-6, p < 0.05), although Cx43 deficient mice were not protected by either IPC or pharmacological preconditioning with diazoxide. Our data demonstrate that modification of RISK and SAFE signaling does not contribute to the role of Cx43 in the increased tolerance to myocardial ischemia-reperfusion injury and in preconditioning protection.


Subject(s)
Connexin 43/deficiency , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Signal Transduction , Adenosine Triphosphate/metabolism , Animals , Cell Death , Connexin 43/genetics , Disease Models, Animal , Energy Metabolism , Enzyme Activation , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Magnetic Resonance Spectroscopy , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Phosphocreatine/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Time Factors , Ventricular Function, Left , Ventricular Pressure
14.
J Cell Mol Med ; 16(8): 1649-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22212640

ABSTRACT

Connexin 43 (Cx43) is present at the sarcolemma and the inner membrane of cardiomyocyte subsarcolemmal mitochondria (SSM). Lack or inhibition of mitochondrial Cx43 is associated with reduced mitochondrial potassium influx, which might affect mitochondrial respiration. Therefore, we analysed the importance of mitochondrial Cx43 for oxygen consumption. Acute inhibition of Cx43 in rat left ventricular (LV) SSM by 18α glycyrrhetinic acid (GA) or Cx43 mimetic peptides (Cx43-MP) reduced ADP-stimulated complex I respiration and ATP generation. Chronic reduction of Cx43 in conditional knockout mice (Cx43(Cre-ER(T)/fl) + 4-OHT, 5-10% of Cx43 protein compared with control Cx43(fl/fl) mitochondria) reduced ADP-stimulated complex I respiration of LV SSM to 47.8 ± 2.4 nmol O(2)/min.*mg protein (n = 8) from 61.9 ± 7.4 nmol O(2)/min.*mg protein in Cx43(fl/fl) mitochondria (n = 10, P < 0.05), while complex II respiration remained unchanged. The LV complex I activities (% of citrate synthase activity) of Cx43(Cre-ER(T)/fl) +4-OHT mice (16.1 ± 0.9%, n = 9) were lower than in Cx43(fl/fl) mice (19.8 ± 1.3%, n = 8, P < 0.05); complex II activities were similar between genotypes. Supporting the importance of Cx43 for respiration, in Cx43-overexpressing HL-1 cardiomyocytes complex I respiration was increased, whereas complex II respiration remained unaffected. Taken together, mitochondrial Cx43 is required for optimal complex I activity and respiration and thus mitochondrial ATP-production.


Subject(s)
Connexin 43/metabolism , Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Oxygen Consumption , Adenosine Triphosphate/biosynthesis , Animals , Connexin 43/antagonists & inhibitors , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Mice , Mitochondria, Heart/drug effects , Oxygen Consumption/drug effects , Peptides/pharmacology , Rats , Rats, Inbred Lew , Sarcolemma/drug effects , Sarcolemma/metabolism
15.
Biochim Biophys Acta ; 1818(8): 1962-70, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21839721

ABSTRACT

Multicellular organisms have developed a variety of mechanisms that allow communication between their cells. Whereas some of these systems, as neurotransmission or hormones, make possible communication between remote areas, direct cell-to-cell communication through specific membrane channels keep in contact neighboring cells. Direct communication between the cytoplasm of adjacent cells is achieved in vertebrates by membrane channels formed by connexins. However, in addition to allowing exchange of ions and small metabolites between the cytoplasms of adjacent cells, connexin channels also communicate the cytosol with the extracellular space, thus enabling a completely different communication system, involving activation of extracellular receptors. Recently, the demonstration of connexin at the inner mitochondrial membrane of cardiomyocytes, probably forming hemichannels, has enlarged the list of actions of connexins. Some of these mechanisms are also shared by a different family of proteins, termed pannexins. Importantly, these systems allow not only communication between healthy cells, but also play an important role during different types of injury. The aim of this review is to discuss the role played by both connexin hemichannels and pannexin channels in cell communication and injury. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Subject(s)
Connexins/metabolism , Gap Junctions/physiology , Heart Injuries/metabolism , Myocardium/pathology , Nerve Tissue Proteins/metabolism , Animals , Cell Communication , Cell Death , Cytoplasm/metabolism , Heart Diseases/metabolism , Humans , Models, Biological , Reperfusion Injury , Signal Transduction
16.
Basic Res Cardiol ; 106(6): 1259-68, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21959501

ABSTRACT

Mitochondrial permeability transition (MPT) is critical in cardiomyocyte death during reperfusion but it is not the only mechanism responsible for cell injury. The objectives of the study is to investigate the role of the duration of myocardial ischemia on mitochondrial integrity and cardiomyocyte death. Mitochondrial membrane potential (ΔΨm, JC-1) and MPT (calcein) were studied in cardiomyocytes from wild-type and cyclophilin D (CyD) KO mice refractory to MPT, submitted to simulated ischemia and 10 min reperfusion. Reperfusion after 15 min simulated ischemia induced a rapid recovery of ΔΨm, extreme cell shortening (contracture) and mitochondrial calcein release, and CyD ablation did not affect these changes or cell death. However, when reperfusion was performed after 25 min simulated ischemia, CyD ablation improved ΔΨm recovery and reduced calcein release and cell death (57.8 ± 4.9% vs. 77.3 ± 4.8%, P < 0.01). In a Langendorff system, CyD ablation increased infarct size after 30 min of ischemia (61.3 ± 6.4% vs. 45.3 ± 4.0%, P = 0.02) but reduced it when ischemia was prolonged to 60 min (52.8 ± 8.1% vs. 87.6 ± 3.7%, P < 0.01). NMR spectroscopy in rat hearts showed a rapid recovery of phosphocreatine after 30 min ischemia followed by a marked decay associated with contracture and LDH release, that were preventable with contractile blockade but not with cyclosporine A. In contrast, after 50 min ischemia, phosphocreatine recovery was impaired even with contractile blockade (65.2 ± 4% at 2 min), and cyclosporine A reduced contracture, LDH release and infarct size (52.1 ± 4.2% vs. 82.8 ± 3.6%, P < 0.01). In conclusion, the duration of ischemia critically determines the importance of MPT on reperfusion injury. Mechanisms other than MPT may play an important role in cell death after less severe ischemia.


Subject(s)
Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Animals , Cell Death , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Magnetic Resonance Spectroscopy , Male , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Knockout , Mitochondria/pathology , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley
17.
Am J Physiol Heart Circ Physiol ; 301(6): H2442-53, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21949115

ABSTRACT

A transient reduction of cell coupling during reperfusion limits myocardial necrosis, but little is known about its arrhythmogenic effects during ischemia-reperfusion. Thus, we analyzed the effect of an extreme reduction in the number of gap junction channels or in their unitary conductance on ventricular arrhythmias during myocardial ischemia-reperfusion. Available gap junction uncouplers have electrophysiological effects independent from their uncoupling actions. Thus, isolated hearts from Cx43(Cre-ER(T)/fl) mice treated with 4-hydroxytamoxifen (4-OHT), from Cx43KI32 mice [in which connexin (Cx)43 was replaced with Cx32], and from control animals were submitted to regional ischemia and reperfusion, and spontaneous and induced ventricular arrhythmias were monitored. In additional hearts, changes in activation time and electrical impedance during global ischemia-reperfusion were assessed. In contrast to treatment with 4-OHT, replacement of Cx43 with Cx32 did not modify baseline activation time or electrical impedance. However, the number of extrasistole and ventricular tachyarrhythmias was higher in isolated hearts from Cx43KI32 and 4-OHT-treated Cx43(Cre-ER(T)/fl) animals versus wild-type animals during normoxia, ischemia (12.29 ± 3.26 and 52.17 ± 22.51 vs. 3.00 ± 1.46 spontaneous tachyarrhythmias, P < 0.05), and reperfusion. The impairment in conduction during ischemia was steeper in isolated hearts from Cx43KI32 animals, whereas changes in myocardial impedance were attenuated during ischemia in both transgenic models, suggesting altered cell-to-cell coupling at baseline. In conclusion, both reduction of Cx43 with 4-OHT and replacement of Cx43 by less-conductive Cx32 were arrhythmogenic under normoxia and ischemia-reperfusion, despite no major effects on baseline electrical properties. These results suggest that modifications in gap junction communication silent under normal conditions may be arrhythmogenic during ischemia-reperfusion.


Subject(s)
Arrhythmias, Cardiac/etiology , Gap Junctions/metabolism , Heart Ventricles/metabolism , Myocardial Reperfusion Injury/complications , Myocardium/metabolism , Action Potentials , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Disease Models, Animal , Down-Regulation , Electric Impedance , Female , Heart Ventricles/physiopathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Perfusion , Permeability , Time Factors , Gap Junction beta-1 Protein
18.
Cardiovasc Res ; 88(1): 30-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20615915

ABSTRACT

Mitochondria are essential for energy supply and cell signalling and may be triggers and effectors of cell death. Mitochondrial respiration is tightly controlled by the matrix Ca(2+) concentration, which is beat-to-beat regulated by uptake and release mainly through the mitochondrial Ca(2+) uniporter and Na(+)/Ca(2+) exchanger, respectively. Recent studies demonstrate that mitochondrial Ca(2+) uptake is more dependent on anatomo-functional microdomains established with the sarcoplasmic reticulum (SR) than on cytosolic Ca(2+). This privileged communication between SR and mitochondria is not restricted to Ca(2+) but may involve ATP and reactive oxygen species, which has important implications in cardiac pathophysiology. The disruption of the SR-mitochondria interaction caused by cell remodelling has been implicated in the deterioration of excitation-contraction coupling of the failing heart. The SR-mitochondria interplay has been suggested to be involved in the depressed Ca(2+) transients and mitochondrial dysfunction observed in diabetic hearts as well as in the genesis of certain arrhythmias, and it may play an important role in myocardial reperfusion injury. During reperfusion, re-energization in the presence of cytosolic Ca(2+) overload results in SR-driven Ca(2+) oscillations that may promote mitochondrial permeability transition (MPT). The relationship between MPT and Ca(2+) oscillations is bidirectional, as recent data show that the induction of MPT in Ca(2+)-overloaded cardiomyocytes may result in mitochondrial Ca(2+) release that aggravates Ca(2+) handling and favours hypercontracture. A more complete characterization of the structural arrangements responsible for SR-mitochondria interplay will allow better understanding of cardiac (patho)physiology but also, and no less important, should serve as a basis for the development of new treatments for cardiac diseases.


Subject(s)
Heart Diseases/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Sarcoplasmic Reticulum/metabolism , Signal Transduction , Animals , Calcium Signaling , Energy Metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Mitochondria, Heart/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardium/pathology , Sarcoplasmic Reticulum/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...