Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 623: 44-50, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35870261

RESUMO

Aging is associated with increased prevalence of life-threatening ventricular arrhythmias, but mechanisms underlying higher susceptibility to arrhythmogenesis and means to prevent such arrhythmias under stress are not fully defined. We aimed to define differences in aging-associated susceptibility to ventricular fibrillation (VF) induction between young and aged hearts. VF induction was attempted in isolated perfused hearts of young (6-month) and aged (24-month-old) male Fischer-344 rats by rapid pacing before and following isoproterenol (1 µM) or global ischemia and reperfusion (I/R) injury with or without pretreatment with low-dose tetrodotoxin, a late sodium current blocker. At baseline, VF could not be induced; however, the susceptibility to inducible VF after isoproterenol and spontaneous VF following I/R was 6-fold and 3-fold higher, respectively, in old hearts (P < 0.05). Old animals had longer epicardial monophasic action potential at 90% repolarization (APD90; P < 0.05) and displayed a loss of isoproterenol-induced shortening of APD90 present in the young. In isolated ventricular cardiomyocytes from older but not younger animals, 4-aminopyridine prolonged APD and induced early afterdepolarizations (EADs) and triggered activity with isoproterenol. Low-dose tetrodotoxin (0.5 µM) significantly shortened APD without altering action potential upstroke and prevented 4-aminopyridine-mediated APD prolongation, EADs, and triggered activity. Tetrodotoxin pretreatment prevented VF induction by pacing in isoproterenol-challenged hearts. Vulnerability to VF following I/R or catecholamine challenge is significantly increased in old hearts that display reduced repolarization reserve and increased propensity to EADs, triggered activity, and ventricular arrhythmogenesis that can be suppressed by low-dose tetrodotoxin, suggesting a role of slow sodium current in promoting arrhythmogenesis with aging.


Assuntos
Arritmias Cardíacas , Fibrilação Ventricular , 4-Aminopiridina/efeitos adversos , Potenciais de Ação/fisiologia , Envelhecimento/fisiologia , Animais , Isoproterenol/efeitos adversos , Masculino , Miócitos Cardíacos , Ratos , Sódio , Tetrodotoxina/farmacologia , Fibrilação Ventricular/tratamento farmacológico , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/prevenção & controle
2.
J Am Heart Assoc ; 10(16): e019948, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34369184

RESUMO

Background Age-related heart diseases are significant contributors to increased morbidity and mortality. Emerging evidence indicates that mitochondria within cardiomyocytes contribute to age-related increased reactive oxygen species (ROS) generation that plays an essential role in aging-associated cardiac diseases. Methods and Results The present study investigated differences between ROS production in cardiomyocytes isolated from adult (6 months) and aged (24 months) Fischer 344 rats, and in cardiac tissue of adult (18-65 years) and elderly (>65 years) patients with preserved cardiac function. Superoxide dismutase inhibitable ferricytochrome c reduction assay (1.32±0.63 versus 0.76±0.31 nMol/mg per minute; P=0.001) superoxide and H2O2 production, measured as dichlorofluorescein diacetate fluorescence (1646±428 versus 699±329, P=0.04), were significantly higher in the aged versus adult cardiomyocytes. Similarity in age-related alteration between rats and humans was identified in mitochondrial-electron transport chain-complex-I-associated increased oxidative-stress by MitoSOX fluorescence (53.66±18.58 versus 22.81±12.60; P=0.03) and in 4-HNE adduct levels (187.54±54.8 versus 47.83±16.7 ng/mg protein, P=0.0063), indicative of increased peroxidation in the elderly. These differences correlated with changes in functional enrichment of genes regulating ROS homeostasis pathways in aged human and rat hearts. Functional merged collective network and pathway enrichment analysis revealed common genes prioritized in human and rat aging-associated networks that underlay enriched functional terms of mitochondrial complex I and common pathways in the aging human and rat heart. Conclusions Aging sensitizes mitochondrial and extramitochondrial mechanisms of ROS buildup within the heart. Network analysis of the transcriptome highlights the critical elements involved with aging-related ROS homeostasis pathways common in rat and human hearts as targets.


Assuntos
Envelhecimento/metabolismo , Metabolismo Energético , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica , Transcriptoma , Adolescente , Adulto , Fatores Etários , Idoso , Envelhecimento/genética , Animais , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Metabolismo Energético/genética , Feminino , Redes Reguladoras de Genes , Humanos , Peroxidação de Lipídeos , Masculino , Pessoa de Meia-Idade , Mitocôndrias Cardíacas/genética , Fosforilação Oxidativa , Estresse Oxidativo/genética , Ratos Endogâmicos F344 , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Adulto Jovem
3.
Biol Open ; 6(3): 326-332, 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28126709

RESUMO

Excessive cardiac fibrosis, characterized by increased collagen-rich extracellular matrix (ECM) deposition, is a major predisposing factor for mechanical and electrical dysfunction in heart failure (HF). The human ventricular fibroblast (hVF) remodeling mechanisms that cause excessive collagen deposition in HF are unclear, although reports suggest a role for intracellular free Ca2+ in fibrosis. Therefore, we determined the association of differences in cellular Ca2+ dynamics and collagen secretion/deposition between hVFs from failing and normal (control) hearts. Histology of left ventricle sections (Masson trichrome) confirmed excessive fibrosis in HF versus normal. In vitro, hVFs from HF showed increased secretion/deposition of soluble collagen in 48 h of culture compared with control [85.9±7.4 µg/106 cells vs 58.5±8.8 µg/106 cells, P<0.05; (Sircol™ assay)]. However, collagen gene expressions (COL1A1 and COL1A2; RT-PCR) were not different. Ca2+ imaging (fluo-3) of isolated hVFs showed no difference in the thapsigargin-induced intracellular Ca2+ release capacity (control 16±1.4% vs HF 17±1.1%); however, Ca2+ influx via store-operated Ca2+ entry/Ca2+ release-activated channels (SOCE/CRAC) was significantly (P≤0.05) greater in HF-hVFs (47±3%) compared with non-failing (35±5%). Immunoblotting for ICRAC channel components showed increased ORAI1 expression in HF-hVFs compared with normal without any difference in STIM1 expression. The Pearson's correlation coefficient for co-localization of STIM1/ORAI1 was significantly (P<0.01) greater in HF (0.5±0.01) than control (0.4±0.01) hVFs. The increase in collagen secretion of HF versus control hVFs was eliminated by incubation of hVFs with YM58483 (10 µM), a selective ICRAC inhibitor, for 48 h (66.78±5.87 µg/106 cells vs 55.81±7.09 µg/106 cells, P=0.27). In conclusion, hVFs from HF have increased collagen secretion capacity versus non-failing hearts and this is related to increase in Ca2+ entry via SOCE and enhanced expression of ORAI, the pore-forming subunit. Therapeutic inhibition of SOCE may reduce the progression of cardiac fibrosis/HF.

4.
Am J Physiol Heart Circ Physiol ; 311(1): H54-63, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27199126

RESUMO

Mitochondria are critical for maintaining normal cardiac function, and a deficit in mitochondrial energetics can lead to the development of the substrate that promotes atrial fibrillation (AF) and its progression. However, the link between mitochondrial dysfunction and AF in humans is still not fully defined. The aim of this study was to elucidate differences in the functional activity of mitochondrial oxidative phosphorylation (OXPHOS) complexes and oxidative stress in right atrial tissue from patients without (non-AF) and with AF (AF) who were undergoing open-heart surgery and were not significantly different for age, sex, major comorbidities, and medications. The overall functional activity of the electron transport chain (ETC), NADH:O2 oxidoreductase activity, was reduced by 30% in atrial tissue from AF compared with non-AF patients. This was predominantly due to a selective reduction in complex I (0.06 ± 0.007 vs. 0.09 ± 0.006 nmol·min(-1)·citrate synthase activity(-1), P = 0.02) and II (0.11 ± 0.012 vs. 0.16 ± 0.012 nmol·min(-1)·citrate synthase activity(-1), P = 0.003) functional activity in AF patients. Conversely, complex V activity was significantly increased in AF patients (0.21 ± 0.027 vs. 0.12 ± 0.01 nmol·min(-1)·citrate synthase activity(-1), P = 0.005). In addition, AF patients exhibited a higher oxidative stress with increased production of mitochondrial superoxide (73 ± 17 vs. 11 ± 2 arbitrary units, P = 0.03) and 4-hydroxynonenal level (77.64 ± 30.2 vs. 9.83 ± 2.83 ng·mg(-1) protein, P = 0.048). Our findings suggest that AF is associated with selective downregulation of ETC activity and increased oxidative stress that can contribute to the progression of the substrate for AF.


Assuntos
Fibrilação Atrial/enzimologia , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Mitocôndrias Cardíacas/enzimologia , Miócitos Cardíacos/enzimologia , Fosforilação Oxidativa , Estresse Oxidativo , Idoso , Idoso de 80 Anos ou mais , Aldeídos/metabolismo , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/cirurgia , Estudos de Casos e Controles , Progressão da Doença , Regulação para Baixo , Feminino , Átrios do Coração/enzimologia , Átrios do Coração/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Superóxidos/metabolismo
5.
PLoS One ; 10(4): e0123046, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25849590

RESUMO

OBJECTIVS: Cytokine-dependent activation of fibroblasts to myofibroblasts, a key event in fibrosis, is accompanied by phenotypic changes with increased secretory and contractile properties dependent on increased energy utilization, yet changes in the energetic profile of these cells are not fully described. We hypothesize that the TGF-ß1-mediated transformation of myofibroblasts is associated with an increase in mitochondrial content and function when compared to naive fibroblasts. METHODS: Cultured NIH/3T3 mouse fibroblasts treated with TGF-ß1, a profibrotic cytokine, or vehicle were assessed for transformation to myofibroblasts (appearance of α-smooth muscle actin [α-SMA] stress fibers) and associated changes in mitochondrial content and functions using laser confocal microscopy, Seahorse respirometry, multi-well plate reader and biochemical protocols. Expression of mitochondrial-specific proteins was determined using western blotting, and the mitochondrial DNA quantified using Mitochondrial DNA isolation kit. RESULTS: Treatment with TGF-ß1 (5 ng/mL) induced transformation of naive fibroblasts into myofibroblasts with a threefold increase in the expression of α-SMA (6.85 ± 0.27 RU) compared to cells not treated with TGF-ß1 (2.52 ± 0.11 RU). TGF-ß1 exposure increased the number of mitochondria in the cells, as monitored by membrane potential sensitive dye tetramethylrhodamine, and expression of mitochondria-specific proteins; voltage-dependent anion channels (0.54 ± 0.05 vs. 0.23 ± 0.05 RU) and adenine nucleotide transporter (0.61 ± 0.11 vs. 0.22 ± 0.05 RU), as well as mitochondrial DNA content (530 ± 12 µg DNA/106 cells vs. 307 ± 9 µg DNA/106 cells in control). TGF-ß1 treatment was associated with an increase in mitochondrial function with a twofold increase in baseline oxygen consumption rate (2.25 ± 0.03 vs. 1.13 ± 0.1 nmol O2/min/106 cells) and FCCP-induced mitochondrial respiration (2.87 ± 0.03 vs. 1.46 ± 0.15 nmol O2/min/106 cells). CONCLUSIONS: TGF-ß1 induced differentiation of fibroblasts is accompanied by energetic remodeling of myofibroblasts with an increase in mitochondrial respiration and mitochondrial content.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Respiração Celular/fisiologia , Fibroblastos/citologia , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Western Blotting , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Técnicas Imunoenzimáticas , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células NIH 3T3 , RNA Mensageiro/metabolismo
6.
PLoS One ; 7(9): e44667, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22973464

RESUMO

OBJECTIVE: Cardiac subsarcolemmal (SSM) and interfibrillar (IFM) mitochondrial subpopulations possess distinct biochemical properties and differ with respect to their protein and lipid compositions, capacities for respiration and protein synthesis, and sensitivity to metabolic challenge, yet their responsiveness to mitochondrially active cardioprotective therapeutics has not been characterized. This study assessed the differential responsiveness of the two mitochondrial subpopulations to diazoxide, a cardioprotective agent targeting mitochondria. METHODS: Mitochondrial subpopulations were freshly isolated from rat ventricles and their morphologies assessed by electron microscopy and enzymatic activities determined using standard biochemical protocols with a plate reader. Oxidative phosphorylation was assessed from State 3 respiration using succinate as a substrate. Calcium dynamics and the status of Ca²âº-dependent mitochondrial permeability transition (MPT) pore and mitochondrial membrane potential were assessed using standard Ca²âº and TPP⁺ ion-selective electrodes. RESULTS: Compared to IFM, isolated SSM exhibited a higher sensitivity to Ca²âº overload-mediated inhibition of adenosine triphosphate (ATP) synthesis with decreased ATP production (from 375±25 to 83±15 nmol ATP/min/mg protein in SSM, and from 875±39 to 583±45 nmol ATP/min/mg protein in IFM). In addition, SSM exhibited reduced Ca²âº-accumulating capacity as compared to IFM (230±13 vs. 450±46 nmol Ca²âº/mg protein in SSM and IFM, respectively), suggestive of increased Ca²âº sensitivity of MPT pore opening. Despite enhanced susceptibility to stress, SSM were more responsive to the protective effect of diazoxide (100 µM) against Ca²âº overload-mediated inhibition of ATP synthesis (67% vs. 2% in SSM and IFM, respectively). CONCLUSION: These results provide evidence for the distinct sensitivity of cardiac SSM and IFM toward Ca²âº-dependent metabolic stress and the protective effect of diazoxide on mitochondrial energetics.


Assuntos
Cardiotônicos/farmacologia , Diazóxido/farmacologia , Mitocôndrias/efeitos dos fármacos , Miocárdio/citologia , Miofibrilas/metabolismo , Sarcolema/metabolismo , Trifosfato de Adenosina/biossíntese , Análise de Variância , Animais , Western Blotting , Cálcio/metabolismo , Citrato (si)-Sintase/metabolismo , Masculino , Microscopia Eletrônica , Mitocôndrias/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
7.
J Biol Chem ; 287(10): 7692-700, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22228763

RESUMO

We proposed previously that closure of voltage-dependent anion channels (VDAC) in the mitochondrial outer membrane after ethanol exposure leads to suppression of mitochondrial metabolite exchange. Because ureagenesis requires extensive mitochondrial metabolite exchange, we characterized the effect of ethanol and its metabolite, acetaldehyde (AcAld), on total and ureagenic respiration in cultured rat hepatocytes. Ureagenic substrates increased cellular respiration from 15.8 ± 0.9 nmol O(2)/min/10(6) cells (base line) to 29.4 ± 1.7 nmol O(2)/min/10(6) cells in about 30 min. Ethanol (0-200 mM) suppressed extra respiration after ureagenic substrates (ureagenic respiration) by up to 51% but not base line respiration. Urea formation also declined proportionately. Inhibition of alcohol dehydrogenase, cytochrome P450 2E1, and catalase with 4-methylpyrazole, trans-1,2-dichloroethylene, and 3-amino-1,2,3-triazole restored ethanol-suppressed ureagenic respiration by 46, 37, and 66%, respectively. By contrast, inhibition of aldehyde dehydrogenase with phenethyl isothiocyanate increased the inhibitory effect of ethanol on ureagenic respiration by an additional 60%. AcAld, an intermediate product of ethanol oxidation, suppressed ureagenic respiration with an apparent IC(50) of 125 µM. AcAld also inhibited entry of 3-kDa rhodamine-conjugated dextran in the mitochondrial intermembrane space of digitonin-permeabilized hepatocytes, indicative of VDAC closure. In conclusion, AcAld, derived from ethanol metabolism, suppresses ureagenesis in hepatocytes mediated by closure of VDAC.


Assuntos
Acetaldeído/metabolismo , Depressores do Sistema Nervoso Central/farmacocinética , Etanol/farmacocinética , Hepatócitos/metabolismo , Mitocôndrias Hepáticas/metabolismo , Ureia/metabolismo , Álcool Desidrogenase/metabolismo , Animais , Catalase/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Citocromo P-450 CYP2E1/metabolismo , Ativadores de Enzimas/farmacologia , Etanol/farmacologia , Masculino , Consumo de Oxigênio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Canais de Ânion Dependentes de Voltagem/metabolismo
8.
Biochim Biophys Acta ; 1818(6): 1536-44, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22172804

RESUMO

Voltage dependent anion channels (VDAC) are highly conserved proteins that are responsible for permeability of the mitochondrial outer membrane to hydrophilic metabolites like ATP, ADP and respiratory substrates. Although previously assumed to remain open, VDAC closure is emerging as an important mechanism for regulation of global mitochondrial metabolism in apoptotic cells and also in cells that are not dying. During hepatic ethanol oxidation to acetaldehyde, VDAC closure suppresses exchange of mitochondrial metabolites, resulting in inhibition of ureagenesis. In vivo, VDAC closure after ethanol occurs coordinately with mitochondrial uncoupling. Since acetaldehyde passes through membranes independently of channels and transporters, VDAC closure and uncoupling together foster selective and more rapid oxidative metabolism of toxic acetaldehyde to nontoxic acetate by mitochondrial aldehyde dehydrogenase. In single reconstituted VDAC, tubulin decreases VDAC conductance, and in HepG2 hepatoma cells, free tubulin negatively modulates mitochondrial membrane potential, an effect enhanced by protein kinase A. Tubulin-dependent closure of VDAC in cancer cells contributes to suppression of mitochondrial metabolism and may underlie the Warburg phenomenon of aerobic glycolysis. This article is part of a Special Issue entitled: VDAC structure, function, and regulation of mitochondrial metabolism.


Assuntos
Etanol/metabolismo , Glicólise , Mitocôndrias/metabolismo , Canais de Ânion Dependentes de Voltagem/metabolismo , Animais , Humanos , Modelos Biológicos , Neoplasias/metabolismo
9.
Nitric Oxide ; 22(1): 30-6, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19914388

RESUMO

We report the therapeutic potential of S-nitroso-N-acetylpenicillamine-derivatized generation-4 polyamidoamine dendrimers (G4-SNAP) for reducing ischemia/reperfusion (I/R) injury in an isolated, perfused rat heart. The use of this dendrimer scaffold to deliver the nitrosothiol therapeutic did not inhibit NO donor activity as the required dose of G4-SNAP to minimize I/R injury (31nM corresponding to 2microM SNAP) was consistent with the optimum concentration of small molecule SNAP alone. An exploration of G4-SNAP NO release kinetics in the presence of physiologically relevant concentrations of glutathione (GSH) indicated enhanced NO release (t[NO]=1.28microM NO/mg) at 500microM GSH. Reperfusion experiments conducted with 500microM GSH further lowered the optimal therapeutic G4-SNAP dose to 230pM (i.e., 15nM SNAP). The unique combination of G4-SNAP dendrimer and glutathione trigger represents a novel strategy with possible clinical relevance toward salvaging ischemic tissue.


Assuntos
Dendrímeros/farmacologia , Glutationa/farmacologia , Óxido Nítrico/metabolismo , Poliaminas/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Células Cultivadas , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo
10.
Mech Ageing Dev ; 129(6): 304-12, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400259

RESUMO

Aging is associated with progressive decline in energetic reserves compromising cardiac performance and tolerance to injury. Although deviations in mitochondrial functions have been documented in senescent heart, the molecular bases for the decline in energy metabolism are only partially understood. Here, high-throughput transcription profiles of genes coding for mitochondrial proteins in ventricles from adult (6-months) and aged (24-months) rats were compared using microarrays. Out of 614 genes encoding for mitochondrial proteins, 94 were differentially expressed with 95% downregulated in the aged. The majority of changes affected genes coding for proteins involved in oxidative phosphorylation (39), substrate metabolism (14) and tricarboxylic acid cycle (6). Compared to adult, gene expression changes in aged hearts translated into a reduced mitochondrial functional capacity, with decreased NADH-dehydrogenase and F(0)F(1) ATPase complex activities and capacity for oxygen-utilization and ATP synthesis. Expression of genes coding for transcription co-activator factors involved in the regulation of mitochondrial metabolism and biogenesis were downregulated in aged ventricles without reduction in mitochondrial density. Thus, aging induces a selective decline in activities of oxidative phosphorylation complexes I and V within a broader transcriptional downregulation of mitochondrial genes, providing a substrate for reduced energetic efficiency associated with senescence.


Assuntos
Envelhecimento , Perfilação da Expressão Gênica , Mitocôndrias/metabolismo , Miocárdio/metabolismo , Transcrição Gênica , Trifosfato de Adenosina/metabolismo , Animais , Senescência Celular , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação Oxidativa , ATPases Translocadoras de Prótons/metabolismo , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Biomacromolecules ; 8(12): 3853-9, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18004811

RESUMO

The cytotoxicity and time-dependent membrane disruption by polypropylenimine dendrimer conjugates on cultured human umbilical vein endothelial cells (HUVEC) is reported. Fluorescently labeled derivatives of generation 5 polypropylenimine dendrimers were prepared via conversion of amines to acetamides or through the covalent attachment of high molecular weight poly(ethylene glycol) (PEG) chains. Direct interactions between the fluorescent dendrimer conjugates and HUVEC were monitored using confocal fluorescence microscopy to track dendrimer movement across the plasma membrane and the fluorescent staining of cell nuclei. Propidium iodide and lactate dehydrogenase cytotoxicity assays confirmed that chemical modification of the surface amines of the parental dendrimer to neutral acetamide or PEG functionalities eliminated their acute cytotoxicity. Cationic primary-amine-containing dendrimers demonstrated drastic time-dependent changes in the plasma membrane permeability and prominent cytotoxicity. However, complete removal of the primary amines or masking of the cationic surface via PEGylation decreased dendrimer cytotoxicity. Thus, preventing electrostatic interactions of dendrimers with cellular membranes apparently is a necessary step toward minimizing the toxicity of delivery vehicles to the endothelium.


Assuntos
Citostáticos/química , Citostáticos/toxicidade , Dendrímeros/química , Dendrímeros/toxicidade , Células Endoteliais/efeitos dos fármacos , Polipropilenos/química , Polipropilenos/toxicidade , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Humanos
12.
J Biol Chem ; 282(12): 8860-72, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17213201

RESUMO

Adaphostin is a dihydroquinone derivative that is undergoing extensive preclinical testing as a potential anticancer drug. Previous studies have suggested that the generation of reactive oxygen species (ROS) plays a critical role in the cytotoxicity of this agent. In this study, we investigated the source of these ROS. Consistent with the known chemical properties of dihydroquinones, adaphostin simultaneously underwent oxidation to the corresponding quinone and generated ROS under aqueous conditions. Interestingly, however, this quinone was not detected in intact cells. Instead, high performance liquid chromatography demonstrated that adaphostin was concentrated by up to 300-fold in cells relative to the extracellular medium and that the highest concentration of adaphostin (3000-fold over extracellular concentrations) was detected in mitochondria. Consistent with a mitochondrial site for adaphostin action, adaphostin-induced ROS production was diminished by >75% in MOLT-4 rho(0) cells, which lack mitochondrial electron transport, relative to parental MOLT-4 cells. In addition, inhibition of oxygen consumption was observed when intact cells were treated with adaphostin. Loading of isolated mitochondria to equivalent adaphostin concentrations caused inhibition of uncoupled oxygen consumption in mitochondria incubated with the complex I substrates pyruvate and malate or the complex II substrate succinate. Further analysis demonstrated that adaphostin had no effect on pyruvate or succinate dehydrogenase activity. Instead, adaphostin inhibited reduced decylubiquinone-induced cytochrome c reduction, identifying complex III as the site of inhibition by this agent. Moreover, adaphostin enhanced the production of ROS by succinate-charged mitochondria. Collectively, these observations demonstrate that mitochondrial respiration rather than direct redox cycling of the hydroquinone moiety is a source of adaphostin-induced ROS and identify complex III as a potential target for antineoplastic agents.


Assuntos
Adamantano/análogos & derivados , Hidroquinonas/farmacologia , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio , Adamantano/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Respiração Celular , DNA-Formamidopirimidina Glicosilase/metabolismo , Transporte de Elétrons , Humanos , Células K562 , Potenciais da Membrana , Oxirredução , Consumo de Oxigênio , Peróxidos/metabolismo , Superóxidos/metabolismo
13.
J Theor Biol ; 243(1): 152-69, 2006 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16859713

RESUMO

Mitochondria play an important role in regulation of Ca2+ homeostasis in a cell. Here we present a mathematical model of mitochondrial ion transport and use this model to analyse different modes of Ca2+ uptake by mitochondria. The model includes transport of H+, Ca2+, K+, inorganic phosphate and oxidative substrates across the inner mitochondrial membrane harboring permeability transition pore (PTP). The detailed description of ion fluxes is based on the experimental ion kinetics in isolated mitochondria. Using the model we show that the kinetics of Ca2+ uptake by mitochondria is regulated by the total amount of Ca2+ in the system and the rate of Ca2+ infusion. Varying these parameters we find three different modes of ion transport. When the total amount of Ca2+ is below 140 nmol Ca2+/mg protein, all available Ca2+ is accumulated in the matrix without activation of the PTP. Between 140 and 160 nmol Ca2+/mg protein, accumulation of Ca2+ generates periodic opening and closure of the PTP and oscillations of ion fluxes. Higher levels of Ca2+ (> 160 nmol Ca2+/mg protein) result in a permanently open PTP, membrane depolarization and loss of small ions from the matrix. We show that in the intermediate range of Ca2+ concentrations the rate of Ca2+ infusion regulates the PTP state, so that slow Ca2+ infusion does not lead to PTP opening, while fast Ca2+ infusion results in an oscillatory state.


Assuntos
Cálcio/metabolismo , Homeostase/fisiologia , Mitocôndrias/metabolismo , Modelos Biológicos , Animais , Relógios Biológicos/fisiologia , Transporte Biológico/fisiologia , Sinalização do Cálcio/fisiologia , Concentração de Íons de Hidrogênio , Potenciais da Membrana/fisiologia , Membranas Mitocondriais/metabolismo , Permeabilidade
14.
Arterioscler Thromb Vasc Biol ; 24(11): 2021-7, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15319267

RESUMO

OBJECTIVE: Endothelial progenitor cells (EPCs) display a unique aptitude to promote angiogenesis and restore endothelial function of injured vessels. How progenitor cells can execute a regenerative program in the unfavorable environment of injury/inflammation-induced oxidative stress is poorly understood. We hypothesized that EPCs are resistant to oxidative stress and that this resistance is due to high expression and activity of antioxidant enzymes. METHODS AND RESULTS: EPCs outgrown from human blood of healthy subjects demonstrated a marked resistance to cytotoxic effect of LY83583 (an generator), tumor necrosis factor-alpha, and serum depletion. LY83583 inhibited in vitro tube formation by human umbilical vein endothelial cells (HUVECs) and human coronary artery endothelial cells (CAECs), but not by EPCs. Compared with HUVECs and CAECs, EPCs exhibited approximately 3- to 4-fold higher expression and activity of manganese superoxide dismutase (MnSOD), but not copper zinc superoxide dismutase (CuZnSOD) or catalase. The antioxidant profile in EPCs was associated with preservation of the mitochondrial network when exposed to LY83583. Moreover, cytotoxic effects of LY83583 on CAECs and HUVECs were reversed by adenoviral overexpression of MnSOD. CONCLUSIONS: Human EPCs are resistant to oxidative stress. High intrinsic expression of MnSOD is a critical mechanism protecting EPCs against oxidative stress.


Assuntos
Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Estresse Oxidativo/fisiologia , Células-Tronco/enzimologia , Células-Tronco/metabolismo , Superóxido Dismutase/biossíntese , Aminoquinolinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Vasos Coronários/citologia , Células Endoteliais/química , Células Endoteliais/efeitos dos fármacos , Genes/genética , Humanos , Mitocôndrias/metabolismo , Fenótipo , Células-Tronco/química , Células-Tronco/efeitos dos fármacos , Superóxido Dismutase/genética , Veias Umbilicais
15.
FEBS Lett ; 568(1-3): 167-70, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15196941

RESUMO

Excessive build-up of mitochondrial protonic potential is harmful to cellular homeostasis, and modulation of inner membrane permeability a proposed countermeasure. Here, we demonstrate that structurally distinct potassium channel openers, diazoxide and pinacidil, facilitated transmembrane proton translocation generating H(+)-selective current through planar phospholipid membrane. Both openers depolarized mitochondria, activated state 4 respiration and reduced oxidative phosphorylation, recapitulating the signature of mitochondrial uncoupling. This effect was maintained in K(+)-free conditions and shared with the prototypic protonophore 2,4-dinitrophenol. Diazoxide, pinacidil and 2,4-dinitrophenol, but not 2,4-dinitrotoluene lacking protonophoric properties, preserved functional recovery of ischemic heart. The identified protonophoric property of potassium channel openers, thus, implicates a previously unrecognized component in their mechanism of cardioprotection.


Assuntos
Cardiotônicos/farmacologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Canais de Potássio/agonistas , Desacopladores/farmacologia , Animais , Diazóxido/farmacologia , Mitocôndrias Cardíacas/fisiologia , Fosforilação Oxidativa , Pinacidil/farmacologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...