Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3528, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664444

RESUMO

Cardiac dysfunction is a hallmark of aging in humans and mice. Here we report that a two-week treatment to restore youthful Bridging Integrator 1 (BIN1) levels in the hearts of 24-month-old mice rejuvenates cardiac function and substantially reverses the aging phenotype. Our data indicate that age-associated overexpression of BIN1 occurs alongside dysregulated endosomal recycling and disrupted trafficking of cardiac CaV1.2 and type 2 ryanodine receptors. These deficiencies affect channel function at rest and their upregulation during acute stress. In vivo echocardiography reveals reduced systolic function in old mice. BIN1 knockdown using an adeno-associated virus serotype 9 packaged shRNA-mBIN1 restores the nanoscale distribution and clustering plasticity of ryanodine receptors and recovers Ca2+ transient amplitudes and cardiac systolic function toward youthful levels. Enhanced systolic function correlates with increased phosphorylation of the myofilament protein cardiac myosin binding protein-C. These results reveal BIN1 knockdown as a novel therapeutic strategy to rejuvenate the aging myocardium.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Envelhecimento , Miocárdio , Proteínas do Tecido Nervoso , Canal de Liberação de Cálcio do Receptor de Rianodina , Proteínas Supressoras de Tumor , Animais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Masculino , Envelhecimento/metabolismo , Camundongos , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Miocárdio/metabolismo , Miocárdio/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Técnicas de Silenciamento de Genes , Endossomos/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Coração/fisiopatologia , Camundongos Endogâmicos C57BL , Humanos , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/genética , Sístole
2.
Anesth Analg ; 106(4): 1049-55, table of contents, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18349172

RESUMO

BACKGROUND: Mitochondrial (m) adenosine triphosphate sensitive potassium (K(ATP)) channel opening has been reported to trigger and/or mediate cardioprotection by volatile anesthetics. However, the effects of volatile anesthetics on mitochondrial function are not well understood. Prevention of mitochondrial matrix volume (MMV) contraction during ischemia may contribute to cardioprotection against ischemia/reperfusion injury. We investigated whether sevoflurane increases MMV and if this increase is mediated by mK(ATP) channel opening. METHODS: Mitochondria from fresh guinea pig hearts were isolated and diluted in buffer that included oligomycin and ATP to inhibit ATP synthesis. Changes in MMV by diazoxide, a known mK(ATP) channel opener, and by different sevoflurane concentrations, were measured by light absorption at 520 nm in the absence or presence of the mK(ATP) channel blocker, 5-hydroxydecanoate. RESULTS: Compared with control, 30-300 microM sevoflurane (approximately 0.2-2.1 vol %) increased MMV by 30%-55%, which was similar to the effect of diazoxide. These increases were blocked by 5-hydroxydecanoate. Higher sevoflurane concentration (1000 microM; 7.1 vol %), however, had no effect on MMV. CONCLUSIONS: In clinically relevant concentrations, sevoflurane increases MMV via mK(ATP) channel opening. Preservation of mitochondrial integrity may contribute to the cardioprotective effects of sevoflurane against ischemia/reperfusion injury. Impaired mitochondrial function at supraclinical anesthetic concentrations may explain the observed biphasic response. These findings add to our understanding of the intracellular mechanisms of volatile anesthetics as cardioprotective drugs.


Assuntos
Éteres Metílicos/farmacologia , Mitocôndrias Cardíacas/ultraestrutura , Trifosfato de Adenosina/metabolismo , Animais , Feminino , Cobaias , Heparina/farmacologia , Cinética , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Sevoflurano , Valinomicina/farmacologia
3.
J Mol Cell Cardiol ; 42(3): 631-42, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17306295

RESUMO

The aim of this study was to investigate the effects of HMR1098, a selective blocker of sarcolemmal ATP-sensitive potassium channel (sarcK(ATP)), in Langendorff-perfused rat hearts submitted to ischemia and reperfusion. The recovery of heart hemodynamic and mitochondrial function, studied on skinned fibers, was analyzed after 30-min global ischemia followed by 20-min reperfusion. Infarct size was quantified on a regional ischemia model after 2-h reperfusion. We report that the perfusion of 10 microM HMR1098 before ischemia, delays the onset of ischemic contracture, improves recovery of cardiac function upon reperfusion, preserves the mitochondrial architecture, and finally decreases infarct size. This HMR1098-induced cardioprotection is prevented by 1 mM 2-mercaptopropionylglycine, an antioxidant, and by 100 nM nifedipine, an L-type calcium channel blocker. Concomitantly, it is shown that HMR1098 perfusion induces (i) a transient and specific inhibition of the respiratory chain complex I and, (ii) an increase in the averaged intracellular calcium concentration probed by the in situ measurement of indo-1 fluorescence. Finally, all the beneficial effects of HMR1098 were strongly inhibited by 5-hydroxydecanoate and abolished by glibenclamide, two mitoK(ATP) blockers. This study demonstrates that the HMR1098-induced cardioprotection occurs indirectly through extracellular calcium influx, respiratory chain complex inhibition, reactive oxygen species production and mitoK(ATP) opening. Taken together, these data suggest that a functional interaction between sarcK(ATP) and mitoK(ATP) exists in isolated rat heart ischemia model, which is mediated by extracellular calcium influx.


Assuntos
Benzamidas/uso terapêutico , Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias Cardíacas/metabolismo , Isquemia Miocárdica/metabolismo , Canais de Potássio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sarcolema/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Bloqueadores dos Canais de Potássio/uso terapêutico , Ratos , Ratos Sprague-Dawley , Sarcolema/efeitos dos fármacos
4.
Am J Physiol Heart Circ Physiol ; 292(3): H1470-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17098831

RESUMO

We showed recently that mitochondrial ATP-dependent K(+) channel (mitoK(ATP)) opening is required for the inotropic response to ouabain. Because mitoK(ATP) opening is also required for most forms of cardioprotection, we investigated whether exposure to ouabain was cardioprotective. We also began to map the signaling pathways linking ouabain binding to Na(+)-K(+)-ATPase with the opening of mitoK(ATP). In Langendorff-perfused rat hearts, 10-80 microM ouabain given before the onset of ischemia resulted in cardioprotection against ischemia-reperfusion injury, as documented by an improved recovery of contractile function and a reduction of infarct size. In skinned cardiac fibers, a ouabain-induced protection of mitochondrial outer membrane integrity, adenine nucleotide compartmentation, and energy transfer efficiency was evidenced by a decreased release of cytochrome c and preserved half-saturation constant of respiration for ADP and adenine nucleotide translocase-mitochondrial creatine kinase coupling, respectively. Ouabain-induced positive inotropy was dose dependent over the range studied, whereas ouabain-induced cardioprotection was maximal at the lowest dose tested. Compared with bradykinin (BK)-induced preconditioning, ouabain was equally efficient. However, the two ligands clearly diverge in the intracellular steps leading to mitoK(ATP) opening from their respective receptors. Thus BK-induced cardioprotection was blocked by inhibitors of cGMP-dependent protein kinase (PKG) or guanylyl cyclase (GC), whereas ouabain-induced protection was not blocked by either agent. Interestingly, however, ouabain-induced inotropy appears to require PKG and GC. Thus 5-hydroxydecanoate (a selective mitoK(ATP) inhibitor), N-(2-mercaptopropionyl)glycine (MPG; a reactive oxygen species scavenger), ODQ (a GC inhibitor), PP2 (a src kinase inhibitor), and KT-5823 (a PKG inhibitor) abolished preconditioning by BK and blocked the inotropic response to ouabain. However, only PP2, 5-HD, and MPG blocked ouabain-induced cardioprotection.


Assuntos
Traumatismo por Reperfusão Miocárdica/prevenção & controle , Ouabaína/uso terapêutico , Canais de Potássio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Quinases da Família src/metabolismo , Difosfato de Adenosina/metabolismo , Animais , Cardiotônicos/uso terapêutico , Creatina/metabolismo , Modelos Animais de Doenças , Frequência Cardíaca/efeitos dos fármacos , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Membranas Mitocondriais/fisiologia , Consumo de Oxigênio , Permeabilidade , Ratos , Ratos Sprague-Dawley
5.
Circ Res ; 99(8): 878-83, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16960097

RESUMO

Mitochondria are key mediators of the cardioprotective signal and the mitochondrial ATP-sensitive K+ channel (mitoK(ATP)) plays a crucial role in originating and transmitting that signal. Recently, protein kinase C epsilon (PKC epsilon) has been identified as a component of the mitoK(ATP) signaling cascade. We hypothesized that PKC epsilon and mitoK(ATP) interact directly to form functional signaling modules in the inner mitochondria membrane. To examine this possibility, we studied K+ flux in liposomes containing partially purified mitoK(ATP). The reconstituted proteins were obtained after detergent extraction of isolated mitochondria, 200-fold purification by ion exchange chromatography, and reconstitution into lipid vesicles. Immunoblot analysis revealed the presence of PKC epsilon in the reconstitutively active fraction. Addition of the PKC activators 12-phorbol 13-myristate acetate, hydrogen peroxide, and the specific PKC epsilon peptide agonist, psi epsilonRACK, each activated mitoK(ATP)-dependent K+ flux in the reconstituted system. This effect of PKC epsilon was prevented by chelerythrine, by the specific PKC epsilon peptide antagonist, epsilonV(1-2), and by the specific mitoK(ATP) inhibitor 5-hydroxydecanoate. In addition, the activating effect of PKC agonists was reversed by exogenous protein phosphatase 2A. These results demonstrate persistent, functional association of mitochondrial PKC epsilon and mitoK(ATP).


Assuntos
Trifosfato de Adenosina/metabolismo , Membranas Mitocondriais/metabolismo , Canais de Potássio/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteolipídeos/metabolismo , Transdução de Sinais/fisiologia , Animais , Ativação Enzimática/fisiologia , Membranas Mitocondriais/enzimologia , Fosfoproteínas Fosfatases/farmacologia , Potássio/metabolismo , Canais de Potássio/isolamento & purificação , Proteína Quinase C-épsilon/isolamento & purificação , Proteína Fosfatase 2 , Proteolipídeos/química , Ratos
6.
Am J Physiol Heart Circ Physiol ; 291(5): H2067-74, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16798828

RESUMO

Opening the mitochondrial ATP-sensitive K(+) channel (mitoK(ATP)) increases levels of reactive oxygen species (ROS) in cardiomyocytes. This increase in ROS is necessary for cardioprotection against ischemia-reperfusion injury; however, the mechanism of mitoK(ATP)-dependent stimulation of ROS production is unknown. We examined ROS production in suspensions of isolated rat heart and liver mitochondria, using fluorescent probes that are sensitive to hydrogen peroxide. When mitochondria were treated with the K(ATP) channel openers diazoxide or cromakalim, their ROS production increased by 40-50%, and this effect was blocked by 5-hydroxydecanoate. ROS production exhibited a biphasic dependence on valinomycin concentration, with peak production occurring at valinomycin concentrations that catalyze about the same K(+) influx as K(ATP) channel openers. ROS production decreased with higher concentrations of valinomycin and with all concentrations of a classical protonophoretic uncoupler. Our studies show that the increase in ROS is due specifically to K(+) influx into the matrix and is mediated by the attendant matrix alkalinization. Myxothiazol stimulated mitoK(ATP)-dependent ROS production, whereas rotenone had no effect. This indicates that the superoxide originates in complex I (NADH:ubiquinone oxidoreductase) of the electron transport chain.


Assuntos
Complexo I de Transporte de Elétrons/fisiologia , Mitocôndrias Cardíacas/fisiologia , Mitocôndrias Hepáticas/fisiologia , Canais de Potássio/fisiologia , Superóxidos/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Antiarrítmicos/farmacologia , Antifúngicos/farmacologia , Cromakalim/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Ácidos Decanoicos/farmacologia , Diazóxido/farmacologia , Corantes Fluorescentes , Concentração de Íons de Hidrogênio , Hidroxiácidos/farmacologia , Ionóforos/farmacologia , Masculino , Metacrilatos/farmacologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Modelos Biológicos , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Tiazóis/farmacologia , Valinomicina/farmacologia , Vasodilatadores/farmacologia
7.
J Biol Chem ; 281(30): 20801-20808, 2006 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-16720572

RESUMO

Myocardial infarction is a manifestation of necrotic cell death as a result of opening of the mitochondrial permeability transition (MPT). Receptor-mediated cardioprotection is triggered by an intracellular signaling pathway that includes phosphatidylinositol 3-kinase, endothelial nitric-oxide synthase, guanylyl cyclase, protein kinase G (PKG), and the mitochondrial K(ATP) channel (mitoK(ATP)). In this study, we explored the pathway that links mitoK(ATP) with the MPT. We confirmed previous findings that diazoxide and activators of PKG or protein kinase C (PKC) inhibited MPT opening. We extended these results and showed that other K(+) channel openers as well as the K(+) ionophore valinomycin also inhibited MPT opening and that this inhibition required reactive oxygen species. By using isoform-specific peptides, we found that the effects of K(ATP) channel openers, PKG, or valinomycin were mediated by a PKCepsilon. Activation of PKCepsilon by phorbol 12-myristate 13-acetate or H(2)O(2) resulted in mitoK(ATP)-independent inhibition of MPT opening, whereas activation of PKCepsilon by PKG or the specific PKCepsilon agonist psiepsilon receptor for activated C kinase caused mitoK(ATP)-dependent inhibition of MPT opening. Exogenous H(2)O(2) inhibited MPT, because of its activation of PKCepsilon, with an IC(50) of 0.4 (+/-0.1) microm. On the basis of these results, we propose that two different PKCepsilon pools regulate this signaling pathway, one in association with mitoK(ATP) and the other in association with MPT.


Assuntos
Trifosfato de Adenosina/química , Peróxido de Hidrogênio/farmacologia , Mitocôndrias/metabolismo , Canais de Potássio/química , Animais , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Permeabilidade , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Distribuição Tecidual
8.
Am J Physiol Heart Circ Physiol ; 290(1): H406-15, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16143645

RESUMO

The mitochondrial ATP-sensitive K+ channel (mitoK(ATP)) has been assigned multiple roles in cell physiology and in cardioprotection. Each of these roles must arise from basic consequences of mitoK(ATP) opening that should be observable at the level of the mitochondrion. MitoK(ATP) opening has been proposed to have three direct effects on mitochondrial physiology: an increase in steady-state matrix volume, respiratory stimulation (uncoupling), and matrix alkalinization. Here, we examine the evidence for these hypotheses through experiments on isolated rat heart mitochondria. Using perturbation techniques, we show that matrix volume is the consequence of a steady-state balance between K+ influx, caused either by mitoK(ATP) opening or valinomycin, and K+ efflux caused by the mitochondrial K+/H+ antiporter. We show that increasing K+ influx with valinomycin uncouples respiration like a classical uncoupler with the important difference that uncoupling via K+ cycling soon causes rupture of the outer mitochondrial membrane and release of cytochrome c. By loading the potassium binding fluorescent indicator into the matrix, we show directly that K+ influx is increased by diazoxide and inhibited by ATP and 5-HD. By loading the fluorescent probe BCECF into the matrix, we show directly that increasing K+ influx with either valinomycin or diazoxide causes matrix alkalinization. Finally, by comparing the effects of mitoK(ATP) openers and blockers with those of valinomycin, we show that four independent assays of mitoK(ATP) activity yield quantitatively identical results for mitoK(ATP)-mediated K+ transport. These results provide decisive support for the hypothesis that mitochondria contain an ATP-sensitive K+ channel and establish the physiological consequences of mitoK(ATP) opening for mitochondria.


Assuntos
Mitocôndrias Cardíacas/fisiologia , Canais de Potássio/fisiologia , Potássio/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Benzofuranos , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Ácidos Decanoicos/farmacologia , Diazóxido/farmacologia , Éteres Cíclicos , Concentração de Íons de Hidrogênio , Hidroxiácidos/farmacologia , Luz , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Dilatação Mitocondrial/efeitos dos fármacos , Oniocompostos/farmacologia , Compostos Organofosforados/farmacologia , Consumo de Oxigênio , Ratos , Ratos Sprague-Dawley , Espalhamento de Radiação , Desacopladores/farmacologia , Valinomicina/farmacologia
9.
Circ Res ; 97(4): 329-36, 2005 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-16037573

RESUMO

Ischemic and pharmacological preconditioning can be triggered by an intracellular signaling pathway in which Gi-coupled surface receptors activate a cascade including phosphatidylinositol 3-kinase, endothelial nitric oxide synthase, guanylyl cyclase, and protein kinase G (PKG). Activated PKG opens mitochondrial KATP channels (mitoKATP) which increase production of reactive oxygen species. Steps between PKG and mitoKATP opening are unknown. We describe effects of adding purified PKG and cGMP on K+ transport in isolated mitochondria. Light scattering and respiration measurements indicate PKG induces opening of mitoKATP similar to KATP channel openers like diazoxide and cromakalim in heart, liver, and brain mitochondria. This effect was blocked by mitoKATP inhibitors 5-hydroxydecanoate, tetraphenylphosphonium, and glibenclamide, PKG-selective inhibitor KT5823, and protein kinase C (PKC) inhibitors chelerythrine, Ro318220, and PKC-epsilon peptide antagonist epsilonV(1-2). MitoKATP are opened by the PKC activator 12-phorbol 13-myristate acetate. We conclude PKG is the terminal cytosolic component of the trigger pathway; it transmits the cardioprotective signal from cytosol to inner mitochondrial membrane by a pathway that includes PKC-epsilon.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Citosol/metabolismo , Precondicionamento Isquêmico Miocárdico , Mitocôndrias Cardíacas/metabolismo , Transdução de Sinais/fisiologia , Trifosfato de Adenosina/farmacologia , Animais , Encéfalo/metabolismo , GMP Cíclico/fisiologia , Masculino , Mitocôndrias Hepáticas/metabolismo , Consumo de Oxigênio , Canais de Potássio/fisiologia , Proteína Quinase C/fisiologia , Ratos , Ratos Wistar , Acetato de Tetradecanoilforbol/farmacologia , Compostos de Tetraetilamônio/farmacologia
10.
J Biol Chem ; 279(31): 32562-8, 2004 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-15138282

RESUMO

The ATP-sensitive potassium channel from the inner mitochondrial membrane (mitoK(ATP)) is a highly selective conductor of K(+) ions. When isolated in the presence of nonionic detergent and reconstituted in liposomes, mitoK(ATP) is inhibited with high affinity by ATP (K((1/2)) = 20-30 microM). We have suggested that holo-mitoK(ATP) is a heteromultimer consisting of an inwardly rectifying K(+) channel (mitoKIR) and a sulfonylurea receptor (Grover, G. J., and Garlid, K. D. (2000) J. Mol. Cell. Cardiol. 32, 677-695). Here, we show that a 55-kDa protein isolated by ethanol extraction and reconstituted in bilayer lipid membranes and liposomes is the mitoKIR. This protein, which lacks the sulfonylurea receptor subunit, is inhibited with low affinity by ATP, with K(1/2) approximately 550 microM. ATP inhibition of both mitoKIR and holo-mitoK(ATP) is reversed by UDP (K((1/2))1/2 = 10-15 microM). Holo-mitoK(ATP) is and diazoxide, and the opened by cromakalim flux through the open channel is inhibited by glibenclamide and 5-hydroxydecanoate. None of these agents has any effect upon mitoKIR. We have identified two compounds that act specifically on mitoKIR. p-diethylaminoethylbenzoate reverses inhibition of mitoKIR by ATP and ADP at micromolar concentrations and also opens mitoK(ATP) in isolated mitochondria. Tetraphenylphosphonium inhibits K(+) flux through both mitoKIR and mitoK(ATP) with the same apparent affinity. These findings support the hypothesis that the 55-kDa mitoKIR is the channel component of mitoK(ATP).


Assuntos
Proteínas de Membrana/química , Canais de Potássio Corretores do Fluxo de Internalização , Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/química , Animais , Benzoatos/farmacologia , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Eletrofisiologia , Íons , Cinética , Bicamadas Lipídicas/metabolismo , Lipossomos/metabolismo , Fígado/metabolismo , Magnésio/química , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Modelos Biológicos , Potássio/química , Potássio/metabolismo , Canais de Potássio/química , Canais de Potássio/metabolismo , Ratos , Receptores de Droga/química , Receptores de Droga/metabolismo , Receptores de Sulfonilureias , Fatores de Tempo , Difosfato de Uridina/metabolismo
11.
Ciênc. cult. (Säo Paulo) ; 48(1/2): 103-8, Jan.-Apr. 1996. ilus, tab
Artigo em Inglês | LILACS | ID: lil-191249

RESUMO

The toxicity of a polyhydroxy derivative of p-benzoquinone, tetrahydroxy-l,4-benzoquinone (THQ), was investigated in Chinese hamster ribroblasts (V79-M8 line). The fast oxidative degradation of THQ, yielding reactive oxygen species, allowed its use as a suitable tool to study the mechanisms of cell injury under oxidative stress. Toxicity of THQ to V79 cells was evaluated by measuring its inhibitory effects on cell growth and upon DNA synthesis rate. Complete prevention of both effects by catalase implicated hydrogen peroxide as the central key in the mechanism of THQ cytotoxicity. The roles of the primary oxidative product of THQ, rhodizonic acid (RDZ), as well as that of calcium, were investigated. The dependence of THQ on hydrogen peroxide for cytotoxicity, together with the possibility of iron chelation by RDZ, led us to propose an intracellular Fenton-type reaction as the mediator of THQ toxicity toward V79 cells. The understanding of THQ toxicity mechanisms can help to gain insights into the way structurally related physiological compounds, such as catechol derivatives, produce their toxic effects on target cells.


Assuntos
Animais , Cricetinae , Benzoquinonas/toxicidade , Espécies Reativas de Oxigênio , Estresse Oxidativo , Divisão Celular , DNA/biossíntese , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Radicais Livres , Peróxido de Hidrogênio , Inibidores da Síntese de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA