Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 198: 221-31, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21884755

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disease in developed countries. The core motor symptoms are attributable to the degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). Why these neurons succumb in PD is not clear. One potential clue has come from the observation that the engagement of L-type Ca²âº channels during autonomous pacemaking elevates the sensitivity of SNc DA neurons to mitochondrial toxins used to create animal models of PD, suggesting that Ca²âº entry is a factor in their selective vulnerability. Recent work has shown that this Ca²âº entry also elevates mitochondrial oxidant stress and that this stress is exacerbated by deletion of DJ-1, a gene associated with an early onset, recessive form of PD. Epidemiological data also support a linkage between L-type Ca²âº channels and the risk of developing PD. This review examines the hypothesis that the primary factor driving neurodegenerative changes in PD is the metabolic stress created by Ca²âº entry, particularly in the face of genetic or environmental factors that compromise oxidative defenses or proteostatic competence.


Assuntos
Cálcio/metabolismo , Neurônios Dopaminérgicos/patologia , Mitocôndrias/metabolismo , Estresse Oxidativo/fisiologia , Doença de Parkinson/patologia , Substância Negra/patologia , Animais , Canais de Cálcio Tipo L/metabolismo , Dopaminérgicos/farmacologia , Dopaminérgicos/uso terapêutico , Neurônios Dopaminérgicos/ultraestrutura , Humanos , Levodopa/farmacologia , Levodopa/uso terapêutico , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico
2.
Free Radic Biol Med ; 47(6): 750-9, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19524665

RESUMO

8-Oxoguanine DNA glycosylase (Ogg1) repairs 8-oxo-7,8-dihydroxyguanine (8-oxoG), one of the most abundant DNA adducts caused by oxidative stress. In the mitochondria, Ogg1 is thought to prevent activation of the intrinsic apoptotic pathway in response to oxidative stress by augmenting DNA repair. However, the predominance of the beta-Ogg1 isoform, which lacks 8-oxoG DNA glycosylase activity, suggests that mitochondrial Ogg1 functions in a role independent of DNA repair. We report here that overexpression of mitochondria-targeted human alpha-hOgg1 (mt-hOgg1) in human lung adenocarcinoma cells with some alveolar epithelial cell characteristics (A549 cells) prevents oxidant-induced mitochondrial dysfunction and apoptosis by preserving mitochondrial aconitase. Importantly, mitochondrial alpha-hOgg1 mutants lacking 8-oxoG DNA repair activity were as effective as wild-type mt-hOgg1 in preventing oxidant-induced caspase-9 activation, reductions in mitochondrial aconitase, and apoptosis, suggesting that the protective effects of mt-hOgg1 occur independent of DNA repair. Notably, wild-type and mutant mt-hOgg1 coprecipitate with mitochondrial aconitase. Furthermore, overexpression of mitochondrial aconitase abolishes oxidant-induced apoptosis whereas hOgg1 silencing using shRNA reduces mitochondrial aconitase and augments apoptosis. These findings suggest a novel mechanism that mt-hOgg1 acts as a mitochondrial aconitase chaperone protein to prevent oxidant-mediated mitochondrial dysfunction and apoptosis that might be important in the molecular events underlying oxidant-induced toxicity.


Assuntos
Adenocarcinoma/enzimologia , DNA Glicosilases/metabolismo , Neoplasias Pulmonares/enzimologia , Mitocôndrias/enzimologia , Proteínas Mutantes/metabolismo , Aconitato Hidratase/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Apoptose/genética , Caspase 9/metabolismo , Linhagem Celular Tumoral , DNA Glicosilases/genética , Reparo do DNA/genética , Células Epiteliais/patologia , Humanos , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas Mutantes/genética , Estresse Oxidativo , Transgenes/genética
3.
Cell Death Differ ; 15(4): 686-90, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18259200

RESUMO

Hypoxia-inducible factor (HIF) is the principal transcription factor involved in the regulation of transcriptional responses to hypoxia. During hypoxia, HIF-alpha levels accumulate and trigger an increase in expression of genes involved in glycolysis, glucose metabolism, mitochondrial function, cell survival, apoptosis, and resistance to oxidative stress. In this regard, HIF activation plays an essential role in triggering cellular protection and metabolic alterations from the consequences of oxygen deprivation. This suggests that HIF activation should confer protection against ischemia-reperfusion (I/R) injury, although this protection might require HIF activation before the onset of lethal ischemia. Studies using enhanced expression of HIF-1alpha suggest that its upregulation may be a beneficial therapeutic modality in the treatment or prevention of ischemic injury. HIF-regulated gene expression may mediate the late phase of preconditioning, and constitutive HIF activity may influence the expression of genes that are required for the cell to be able to respond to acute preconditioning. This article reviews the current literature on the role of HIF in balancing protection and cell death in the face of ischemia and I/R injury.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Transdução de Sinais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular , Sobrevivência Celular , Indução Enzimática , Regulação da Expressão Gênica , Heme Oxigenase-1/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Precondicionamento Isquêmico Miocárdico , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Miocárdio/patologia , Óxido Nítrico Sintase Tipo II/biossíntese , Transdução de Sinais/genética
4.
Cell Biol Toxicol ; 22(3): 149-58, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16555001

RESUMO

Grape seed proanthocyanidin extract (GPSE) at high doses has been shown to exhibit cytotoxicity that is associated with increased apoptotic cell death. Nitric oxide (NO), being a regulator of apoptosis, can be increased in production by the administration of GSPE. In a chick cardiomyocyte study, we demonstrated that high-dose (500 microg/ml) GSPE produces a significantly high level of NO that contributes to increased apoptotic cell death detected by propidium iodide and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining. It is also associated with the depletion of intracellular glutathione (GSH), probably due to increased consumption by NO with the formation of S-nitrosoglutathione. Co-treatment with L-NAME, a NO synthase inhibitor, results in reduction of NO and apoptotic cell death. The decline in reduced GSH/oxidized GSH (GSSG) ratio is also reversed. N-Acetylcysteine, a thiol compound that reacts directly with NO, can reduce the increased NO generation and reverse the decreased GSH/GSSG ratio, thereby attenuating the cytotoxicity induced by high-dose GSPE. Taken together, these results suggest that endogenous NO synthase (NOS) activation and excessive NO production play a key role in the pathogenesis of high-dose GSPE-induced cytotoxicity.


Assuntos
Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico/fisiologia , Extratos Vegetais/toxicidade , Proantocianidinas/toxicidade , Acetilcisteína/farmacologia , Animais , Morte Celular , Sobrevivência Celular , Células Cultivadas , Embrião de Galinha , Relação Dose-Resposta a Droga , Interações Medicamentosas , Glutationa/metabolismo , Extrato de Sementes de Uva
5.
J Biol Chem ; 276(46): 42728-36, 2001 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-11559697

RESUMO

Members of the TNFR (tumor necrosis factor receptor) superfamily are involved in regulating activation and differentiation of cells as well as cell survival and programmed cell death/apoptosis. Multimerization of TNFRs can lead to recruitment of TRAFs (TNFR-associated factors) by the receptors resulting in activation of kinases and transcription factors, such as c-Jun N-terminal kinase and nuclear factor kappaB (NF-kappaB). Signal transduction triggered by TNF-alpha also induces an increase in intracellular reactive oxygen species (ROS). ROS have been suggested to play a role in NF-kappaB activation, which is thought to promote cell survival. However, oxidation of proteins and lipids by ROS can also result in apoptosis. The processes generating intracellular ROS and the mechanism(s) regulating the cellular redox status have not been fully elucidated. We investigated whether TRAFs play a role in controlling intracellular ROS levels. Our results indicate that recruitment of TRAFs to the plasma membrane of human embryonic kidney (HEK) 293 cells is crucial for activation of signaling pathways, which regulate ROS production in mitochondria. TRAF-mediated changes in ROS levels enhanced NF-kappaB activation but were not dependent on NF-kappaB-inducing kinase. Consistent with its anti-apoptotic function, Bcl-x(L) interfered with TRAF-mediated ROS generation but not NF-kappaB activation. Taken together, our results suggest a novel role of TRAFs in signal transduction pathways triggered by TNFR-related proteins, which balance cell survival and apoptosis by regulating the electron transport in mitochondria.


Assuntos
Proteínas de Bactérias/metabolismo , Espécies Reativas de Oxigênio , Transdução de Sinais , Apoptose , Western Blotting , Antígenos CD28/metabolismo , Linhagem Celular , Membrana Celular , Sobrevivência Celular , Citoplasma/metabolismo , Transporte de Elétrons , Ativação Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Antígeno Ki-1/metabolismo , Luciferases/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Oxirredução , Oxigênio/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Frações Subcelulares , Transfecção , Proteína bcl-X
6.
Circ Res ; 88(12): 1259-66, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11420302

RESUMO

We tested whether mitochondria function as the O(2) sensor underlying hypoxic pulmonary vasoconstriction (HPV). In buffer-perfused rat lungs, rotenone, myxothiazol, and diphenyleneiodonium, which inhibit mitochondria in the proximal region of the electron transport chain (ETC), abolished HPV without attenuating the response to U46619. Cyanide and antimycin A inhibit electron transfer in the distal region of the ETC, but they did not abolish HPV. Cultured pulmonary artery (PA) myocytes contract in response to hypoxia or to U46619. The hypoxic response was abolished while the response to U46619 was maintained in mutant (rho(0)) PA myocytes lacking a mitochondrial ETC. To test whether reactive oxygen species (ROS) derived from mitochondria act as signaling agents in HPV, the antioxidants pyrrolidinedithiocarbamate and ebselen and the Cu,Zn superoxide dismutase inhibitor diethyldithiocarbamate were used. These abolished HPV without affecting contraction to U46619, suggesting that ROS act as second messengers. In cultured PA myocytes, oxidation of intracellular 2',7'-dichlorofluorescin diacetate (DCFH) dye increased under 2% O(2), indicating that myocytes increase their generation of H(2)O(2) during hypoxia. This was attenuated by myxothiazol, implicating mitochondria as the source of increased ROS during HPV. These results indicate that mitochondrial ATP is not required for HPV, that mitochondria function as O(2) sensors during hypoxia, and that ROS generated in the proximal region of the ETC act as second messengers in the response.


Assuntos
Hipóxia/metabolismo , Pulmão/irrigação sanguínea , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Vasoconstrição/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Antimicina A/farmacologia , Antioxidantes/farmacologia , Células Cultivadas , Transporte de Elétrons/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Canais Iônicos/efeitos dos fármacos , Metacrilatos , Mitocôndrias/efeitos dos fármacos , Modelos Biológicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Oniocompostos/farmacologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Rotenona/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tiazóis/farmacologia , Desacopladores/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
7.
Am J Physiol Heart Circ Physiol ; 280(3): H1249-55, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11179070

RESUMO

The objective of this study was to examine the role of oxygen radicals, protein kinase C (PKC), and ATP-sensitive K(+) (K(ATP)) channels in mediating flumazenil-produced preconditioning. Chick cardiomyocyte death was quantified using propidium iodide, and oxygen radical generation was assessed using 2',7'-dichlorofluorescin oxidation. Preconditioning was initiated with 10 min of ischemia followed by 10 min of reoxygenation. Alternatively, flumazenil was infused for 10 min and removed 10 min before ischemia. Flumazenil (10 microM) and preconditioning increased oxygen radicals [1,693 +/- 101 (n = 3) and 1,567 +/- 98 (n = 3), respectively, vs. 345 +/- 53 (n = 3) in control] and reduced cell death similarly [22 +/- 3% (n = 5) and 18 +/- 2% (n = 6), respectively, vs. controls 49 +/- 5% (n = 8)]. Protection and increased oxygen radicals by flumazenil were abolished by pretreatment with the antioxidant thiol reductant 2-mercaptopropionyl glycine (800 microM; 52 +/- 10%, n = 6). Specific PKC inhibitors Go-6976 (0.1 microM) and chelerythrine (2 microM), given during ischemia and reoxygenation, blocked flumazenil-produced protection (47 +/- 5%, n = 6). The PKC activator phorbol 12-myristate 13-acetate (0.2 microM), given during ischemia and reoxygenation, reduced cell death similarly to that with flumazenil [17 +/- 4% (n = 6) and 22 +/- 3% (n = 5)]. Finally, 5-hydroxydecanoate (1 mM), a selective mitochondrial K(ATP) channel antagonist given during ischemia and reoxygenation, abolished the protection of flumazenil and phorbol 12-myristate 13-acetate. Thus flumazenil mimics preconditioning to reduce cell death in cardiomyocytes. Oxygen radicals activate mitochondrial K(ATP) channels via PKC during the process.


Assuntos
Flumazenil/farmacologia , Moduladores GABAérgicos/farmacologia , Precondicionamento Isquêmico Miocárdico , Fibras Musculares Esqueléticas/fisiologia , Miocárdio/citologia , Transdução de Sinais/efeitos dos fármacos , Animais , Carbazóis/farmacologia , Morte Celular/efeitos dos fármacos , Embrião de Galinha , Citosol/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/metabolismo , Indóis/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/fisiopatologia , Canais de Potássio/fisiologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de GABA/fisiologia , Tiopronina/farmacologia
8.
J Ethnopharmacol ; 74(1): 63-8, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11137349

RESUMO

Qian-Kun-Nin is a Chinese herbal medicine formulation used for several indications, including the treatment of cardiovascular diseases. This formulation contains herbs which possess antioxidant properties. In this study, Qian-Kun-Nin's ability to confer protection to cardiomyocytes against reactive oxygen species (ROS) generated during mitochondrial electron transport inhibition was tested. The intracellular fluorescent probe 2',7'-dichlorofluorescin diacetate (DCFH-DA, sensitive to H(2)O(2) and hydroxyl radicals) was used to assess intracellular ROS, and propidium iodide (PI) was used to assess viability in cultured chick embryonic cardiomyocytes. Qian-Kun-Nin significantly attenuated oxidation of DCFH in cells exposed to the mitochondrial site III inhibitor, antimycin A, consistent with a decrease in oxidative stress. These attenuated oxidant levels were associated with improved cell survival. After antimycin A exposure, Qian-Kun-Nin decreased cell death from 51. 6+/-3.3% in untreated cells to 27.3+/-3.8% in treated cells at 2 h. We conclude that Qian-Kun-Nin attenuates oxidant stress and protects cells from lethal oxidant damage during mitochondrial electron transport inhibition, and thus its therapeutic potential in treating cardiovascular diseases may relate to its antioxidant properties.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Medicina Tradicional Chinesa , Mitocôndrias Cardíacas/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Análise de Variância , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Medicamentos de Ervas Chinesas/isolamento & purificação , Transporte de Elétrons/efeitos dos fármacos , Etnofarmacologia , Espécies Reativas de Oxigênio
9.
Am J Pathol ; 157(2): 679-88, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10934170

RESUMO

TRAF4 is one of six identified members of the family of TNFR-associated factors. While the other family members have been found to play important roles in the development and maintenance of a normal immune system, the importance of TRAF4 has remained unclear. To address this issue, we have generated TRAF4-deficient mice. Despite widespread expression of TRAF4 in the developing embryo, as well as in the adult, lack of TRAF4 expression results in a localized, developmental defect of the upper respiratory tract. TRAF4-deficient mice are born with a constricted upper trachea at the site of the tracheal junction with the larynx. This narrowing of the proximal end of the trachea results in respiratory air flow abnormalities and increases rates of pulmonary inflammation. These data demonstrate that TRAF4 is required to regulate the anastomosis of the upper and lower respiratory systems during development.


Assuntos
Proteínas/metabolismo , Traqueia/anormalidades , Animais , Modelos Animais de Doenças , Marcação de Genes , Humanos , Laringe/embriologia , Laringe/fisiopatologia , Pulmão/embriologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Proteínas/genética , Transtornos Respiratórios/genética , Transtornos Respiratórios/fisiopatologia , Fator 4 Associado a Receptor de TNF , Traqueia/embriologia , Traqueia/fisiopatologia , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral
10.
Oncogene ; 19(34): 3840-8, 2000 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-10951577

RESUMO

The transcription factor p53 can induce growth arrest or death in cells. Tumor cells that develop mutations in p53 demonstrate a diminished apoptotic potential, which may contribute to growth and tumor metastasis. Cellular levels of p53 are stabilized during hypoxia. The present study tested the hypothesis that reactive oxygen species (ROS) released from mitochondria regulate the cytosolic redox state and are required for the stabilization of p53 protein levels in response to hypoxia. Our results indicate that hypoxia (1.5% O2) increases mitochondrial ROS generation and increases p53 protein levels in human breast carcinoma MCF-7 cells and in normal human diploid fibroblast IMR-90 cells. MCF-7 cells depleted of their mitochondrial DNA (rho(o) cells) failed to stabilize p53 protein levels during hypoxia. The antioxidant N-acetylcysteine and the Cu/Zn superoxide dismutase inhibitor diethyldithiocarbamic acid abolished the hypoxia-induced increases in ROS and p53 levels. Rotenone, an inhibitor of mitochondrial complex I, and 4,4'-diisothiocyanato-stilbene-2,2'-disulfonate, a mitochondrial anion channel inhibitor, also abolished the increase in ROS signal and p53 levels during hypoxia. The p53-dependent gene p21WAF1/CIP1 was also induced by hypoxia in both MCF-7 and IMR-90 cells without affecting the growth rate of either cell line. In contrast, both cell lines exhibited increases in p21WAF1/CIP1 expression and growth arrest after gamma irradiation. Primary chick cardiac myocytes and murine embryonic fibroblasts also showed an increase in p53 protein levels in response to hypoxia without cell death or growth arrest. These results indicate that mitochondria regulate p53 protein levels during hypoxia through a redox-dependent mechanism involving ROS. Despite p53-induction, hypoxia alone does not cause either growth arrest or cell death.


Assuntos
Hipóxia Celular , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Ciclo Celular/efeitos da radiação , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Células Cultivadas , Embrião de Galinha , Cobalto/farmacologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Ditiocarb/farmacologia , Fibroblastos , Fluoresceínas/análise , Fluoresceínas/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miocárdio/citologia , Rotenona/farmacologia , Superóxido Dismutase/antagonistas & inibidores , Células Tumorais Cultivadas , Desacopladores/farmacologia
11.
J Immunol ; 165(2): 1013-21, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10878378

RESUMO

The transcription factor NF-kappa B stimulates the transcription of proinflammatory cytokines including TNF-alpha. LPS (endotoxin) and hypoxia both induce NF-kappa B activation and TNF-alpha gene transcription. Furthermore, hypoxia augments LPS induction of TNF-alpha mRNA. Previous reports have indicated that antioxidants abolish NF-kappa B activation in response to LPS or hypoxia, which suggests that reactive oxygen species (ROS) are involved in NF-kappa B activation. This study tested whether mitochondrial ROS are required for both NF-kappaB activation and the increase in TNF-alpha mRNA levels during hypoxia and LPS. Our results indicate that hypoxia (1.5% O2) stimulates NF-kappa B and TNF-alpha gene transcription and increases ROS generation as measured by the oxidant sensitive dye 2',7'-dichlorofluorescein diacetate in murine macrophage J774.1 cells. The antioxidants N-acetylcysteine and pyrrolidinedithiocarbamic acid abolished the hypoxic activation of NF-kappa B, TNF-alpha gene transcription, and increases in ROS levels. Rotenone, an inhibitor of mitochondrial complex I, abolished the increase in ROS signal, the activation of NF-kappa B, and TNF-alpha gene transcription during hypoxia. LPS stimulated NF-kappa B and TNF-alpha gene transcription but not ROS generation in J774.1 cells. Rotenone, pyrrolidinedithiocarbamic acid, and N-acetylcysteine had no effect on the LPS stimulation of NF-kappa B and TNF-alpha gene transcription, indicating that LPS activates NF-kappa B and TNF-alpha gene transcription through a ROS-independent mechanism. These results indicate that mitochondrial ROS are required for the hypoxic activation of NF-kappa B and TNF-alpha gene transcription, but not for the LPS activation of NF-kappa B.


Assuntos
Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Oxidantes/fisiologia , Transcrição Gênica/imunologia , Fator de Necrose Tumoral alfa/genética , Animais , Hipóxia Celular/genética , Hipóxia Celular/imunologia , Linhagem Celular , DNA/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Ligação Proteica/imunologia , Biossíntese de Proteínas , RNA Mensageiro/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
12.
J Biol Chem ; 275(33): 25130-8, 2000 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-10833514

RESUMO

During hypoxia, hypoxia-inducible factor-1alpha (HIF-1alpha) is required for induction of a variety of genes including erythropoietin and vascular endothelial growth factor. Hypoxia increases mitochondrial reactive oxygen species (ROS) generation at Complex III, which causes accumulation of HIF-1alpha protein responsible for initiating expression of a luciferase reporter construct under the control of a hypoxic response element. This response is lost in cells depleted of mitochondrial DNA (rho(0) cells). Overexpression of catalase abolishes hypoxic response element-luciferase expression during hypoxia. Exogenous H(2)O(2) stabilizes HIF-1alpha protein during normoxia and activates luciferase expression in wild-type and rho(0) cells. Isolated mitochondria increase ROS generation during hypoxia, as does the bacterium Paracoccus denitrificans. These findings reveal that mitochondria-derived ROS are both required and sufficient to initiate HIF-1alpha stabilization during hypoxia.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/química , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Hipóxia , Mitocôndrias/metabolismo , Proteínas Nucleares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição , Androstadienos/farmacologia , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Quelantes/farmacologia , Cobalto/farmacologia , Citosol/química , Desferroxamina/farmacologia , Relação Dose-Resposta a Droga , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Inibidores Enzimáticos/farmacologia , Genes Reporter , Humanos , Peróxido de Hidrogênio/metabolismo , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Immunoblotting , Toxinas Marinhas , Mitocôndrias/enzimologia , Mitocôndrias Hepáticas/metabolismo , Modelos Biológicos , Oxazóis/farmacologia , Oxirredução , Oxigênio/metabolismo , Paracoccus denitrificans/metabolismo , Ratos , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas , Wortmanina
13.
J Appl Physiol (1985) ; 88(5): 1880-9, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10797153

RESUMO

Hypoxia elicits a variety of adaptive responses at the tissue level, at the cellular level, and at the molecular level. A physiological response to hypoxia requires the existence of an O(2) sensor coupled to a signal transduction system, which in turn activates the functional response. Although much has been learned about the signaling systems activated by hypoxia, no consensus exists regarding the nature of the underlying O(2) sensor or whether multiple sensors exist. Among previously considered mechanisms, heme proteins have been suggested to undergo allosteric modification in response to O(2) binding or release at different PO(2) levels. Other studies suggest that ion channels may change conductance as a function of PO(2), allowing them to signal the onset of hypoxia. Still other studies suggest that NADPH oxidase may decrease its generation of reactive O(2) species (ROS) during hypoxia. Recent data suggest that mitochondria may function as O(2) sensors by increasing their generation of ROS during hypoxia. These oxidant signals appear to act as second messengers in the adaptive responses to hypoxia in a variety of cell types. Such observations contribute to a growing awareness that mitochondria do more than just generate ATP, in that they initiate signaling cascades involved in adaptive responses to hypoxia and that they participate in the control of cell death pathways.


Assuntos
Mitocôndrias/fisiologia , Oxigênio/metabolismo , Animais , Hemeproteínas/fisiologia , Humanos , Canais Iônicos/fisiologia , Mitocôndrias/metabolismo , Modelos Biológicos , NADP/fisiologia , Espécies Reativas de Oxigênio/fisiologia
14.
Proc Natl Acad Sci U S A ; 97(9): 4666-71, 2000 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-10781072

RESUMO

Coupled cellular respiration requires that ATP and ADP be efficiently exchanged between the cytosol and the mitochondrial matrix. When growth factors are withdrawn from dependent cells, metabolism is disrupted by a defect in ATP/ADP exchange across the mitochondrial membranes. Unexpectedly, we find that this defect results from loss of outer mitochondrial membrane permeability to metabolic anions. This decrease in anion permeability correlates with the changes in conductance properties that accompany closure of the voltage-dependent anion channel (also known as mitochondrial porin). Loss of outer membrane permeability (i) results in the accumulation of stored metabolic energy within the intermembrane space in the form of creatine phosphate, (ii) is prevented by the outer mitochondrial membrane proteins Bcl-x(L) and Bcl-2, and (iii) can be reversed by growth factor readdition. If outer membrane impermeability persists, the disruption of mitochondrial homeostasis culminates in loss of outer mitochondrial membrane integrity, cytochrome c redistribution, and apoptosis. The recognition that outer membrane permeability is regulated under physiological conditions has important implications for the understanding of bioenergetics and cell survival.


Assuntos
Sobrevivência Celular , Membranas Intracelulares/fisiologia , Mitocôndrias/metabolismo , Consumo de Oxigênio , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Divisão Celular , Linhagem Celular , Creatina Quinase/metabolismo , Cinética , Camundongos , Permeabilidade , Fosfocreatina/metabolismo , Porinas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Canais de Ânion Dependentes de Voltagem , Proteína bcl-X
15.
Circ Res ; 86(5): 541-8, 2000 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-10720416

RESUMO

Cardiomyocyte death after ischemia/reperfusion correlates with oxidant stress, and antioxidants confer protection in that model. Preconditioning (PC) with hypoxia or adenosine also confers protection, leading us to hypothesize that PC protects by attenuating oxidant generation during subsequent ischemia/reperfusion. Chick cardiomyocytes were preconditioned with 10 minutes of hypoxia or adenosine (100 micromol/L), followed by 1 hour of simulated ischemia and 3 hours of reperfusion. Adenosine PC decreased cell death from 50+/-3% to 18+/-4% and enhanced the return of contractions during reperfusion, as observed previously with hypoxic PC. A transient burst of dichlorofluorescein (sensitive to H2O2 oxidation that was significantly attenuated by PC initiated by hypoxia or adenosine was seen at reperfusion. The protein kinase C (PKC) inhibitor Go-6976 and the mitochondrial ATP-sensitive K(+) (K(ATP)) channel inhibitor 5-hydroxydecanoate each abolished protection and abrogated the PC-induced attenuation of reperfusion oxidant stress. By contrast, when given only at reperfusion, the K(+) channel opener pinacidil or the antioxidants 2-mercaptopropionylglycine and 1,10-phenanthroline decreased oxidant stress at reperfusion and improved survival and return of contractions. Thus, PC protection is associated with an attenuation of the oxidant burst at reperfusion, regardless of the method by which PC is triggered. Loss of PC protection associated with PKC inhibition or K(ATP) channel inhibitors is associated with a restoration of that oxidant stress. These results suggest a mechanism for PC protection and reveal a functional link between PKC activation and K(ATP) channel activation in that pathway.


Assuntos
Precondicionamento Isquêmico , Isquemia Miocárdica/metabolismo , Miocárdio/enzimologia , Estresse Oxidativo/fisiologia , Traumatismo por Reperfusão/metabolismo , Animais , Antiarrítmicos/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Embrião de Galinha , Ácidos Decanoicos/farmacologia , Peróxido de Hidrogênio/farmacologia , Hidroxiácidos/farmacologia , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/enzimologia , Contração Miocárdica/fisiologia , Miocárdio/química , Miocárdio/citologia , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Pinacidil/farmacologia , Canais de Potássio/fisiologia , Proteína Quinase C/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Vasodilatadores/farmacologia
16.
Am J Physiol ; 277(6): H2240-6, 1999 12.
Artigo em Inglês | MEDLINE | ID: mdl-10600842

RESUMO

Although a burst of oxidants has been well described with reperfusion, less is known about the oxidants generated by the highly reduced redox state and low O(2) of ischemia. This study aimed to further identify the species and source of these oxidants. Cardiomyocytes were exposed to 1 h of simulated ischemia while oxidant generation was assessed by intracellular dihydroethidine (DHE) oxidation. Ischemia increased DHE oxidation significantly (0.7 +/- 0.1 to 2.3 +/- 0.3) after 1 h. Myxothiazol (mitochondrial site III inhibitor) attenuated oxidation to 1.3 +/- 0.1, as did the site I inhibitors rotenone (1.0 +/- 0.1), amytal (1.1 +/- 0.1), and the flavoprotein oxidase inhibitor diphenyleneiodonium (0.9 +/- 0.1). By contrast, the site IV inhibitor cyanide, as well as inhibitors of xanthine oxidase (allopurinol), nitric oxide synthase (nitro-L-arginine methyl ester), and NADPH oxidase (apocynin), had no effect. Finally, DHE oxidation increased with Cu- and Zn-containing superoxide dismutase (SOD) inhibition using diethyldithiocarbamate (2.7 +/- 0.1) and decreased with exogenous SOD (1.1 +/- 0.1). We conclude that significant superoxide generation occurs during ischemia before reperfusion from the ubisemiquinone site of the mitochondrial electron transport chain.


Assuntos
Mitocôndrias Cardíacas/metabolismo , Isquemia Miocárdica/metabolismo , Reperfusão Miocárdica , Miocárdio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Acetofenonas/farmacologia , Alopurinol/farmacologia , Animais , Células Cultivadas , Embrião de Galinha , Citosol/enzimologia , Inibidores Enzimáticos/farmacologia , Coração/efeitos dos fármacos , Cinética , Metacrilatos , Mitocôndrias Cardíacas/efeitos dos fármacos , Miocárdio/citologia , NG-Nitroarginina Metil Éster/farmacologia , Oxirredução , Rotenona/farmacologia , Superóxido Dismutase/metabolismo , Tiazóis/farmacologia
17.
Am J Physiol ; 277(6): H2504-9, 1999 12.
Artigo em Inglês | MEDLINE | ID: mdl-10600875

RESUMO

We examined the ability of ACh to mimic ischemic preconditioning in cardiomyocytes and the role of ATP-sensitive potassium (KATP) channels and mitochondrial reactive oxygen species (ROS) in mediating this effect. Chick embryonic ventricular myocytes were studied in a flow-through chamber while flow rate, pH, PO2, and PCO2 were controlled. Cell viability was quantified with propidium iodide (5 microM), and production of ROS was measured using 2', 7'-dichlorofluorescin diacetate. Data were expressed as means +/- SE. Preconditioning with 10 min of ischemia followed by 10 min of reoxygenation or 10 min of ACh (1 mM) followed by a drug-free period before 1 h of ischemia and 3 h of reoxygenation reduced cell death to the same extent [preconditioning 19 +/- 2% (n = 6, P < 0.05) ACh 21 +/- 5% (n = 6, P < 0.05) vs controls 42 +/- 5% (n = 9)]. Like preconditioning, ACh increased ROS production threefold before ischemia [0.60 +/- 0.16 (n = 7, P < 0.05) vs. controls, 0.16 +/- 0. 03 (n = 6); arbitrary units]. Protection and increased ROS production during ACh preconditioning were abolished with 5-hydroxydecanoate (5-HD, 100 microM), a selective mitochondrial K(ATP) channel antagonist, and the thiol reductant 2-mercaptopropionyl glycine (2-MPG, 1 mM), an antioxidant [cell death: 5-HD+ACh 37 +/- 7% (n = 5), 2-MPG+ACh 47 +/- 6% (n = 6); ROS signals: 5-HD+ACh 0.09 +/- 0.03 (n = 5), 2-MPG+ACh 0.01 +/- 0.04 (n = 4)]. In addition, ACh-induced ROS signaling was blocked by the mitochondrial site III electron transport inhibitor myxothiazol (0.02 +/- 0.07, n = 5). These results demonstrate that activation of mitochondrial K(ATP) channels and increased ROS production from mitochondria are important intracellular signals that participate in ACh-induced preconditioning in cardiomyocytes.


Assuntos
Acetilcolina/farmacologia , Coração/fisiologia , Precondicionamento Isquêmico/métodos , Espécies Reativas de Oxigênio/fisiologia , Animais , Antiarrítmicos/farmacologia , Cardiotônicos/farmacologia , Células Cultivadas , Embrião de Galinha , Ácidos Decanoicos/farmacologia , Transporte de Elétrons/efeitos dos fármacos , Coração/efeitos dos fármacos , Ventrículos do Coração/embriologia , Concentração de Íons de Hidrogênio , Hidroxiácidos/farmacologia , Metacrilatos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Isquemia Miocárdica , Reperfusão Miocárdica , Miocárdio/citologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Transdução de Sinais , Tiazóis/farmacologia , Fatores de Tempo , Tiopronina/farmacologia
18.
Am J Physiol ; 277(5): L1057-65, 1999 11.
Artigo em Inglês | MEDLINE | ID: mdl-10564193

RESUMO

Prolonged hypoxia produces reversible changes in endothelial permeability, but the mechanisms involved are not fully known. Previous studies have implicated reactive oxygen species (ROS) and cytokines in the regulation of permeability. We tested whether prolonged hypoxia alters permeability to increasing ROS generation, which amplifies cytokine production. Human umbilical vein endothelial cell (HUVEC) monolayers were exposed to hypoxia while secretion of tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1alpha, IL-6, and IL-8 was measured. IL-6 and IL-8 secretion increased fourfold over 24 h in a pattern corresponding to changes in HUVEC permeability measured by transendothelial electrical resistance (TEER). Addition of exogenous IL-6 to normoxic HUVEC monolayers caused time-dependent changes in TEER that mimicked the hypoxic response. An antibody to IL-6 significantly attenuated the hypoxia-induced changes in TEER (86 +/- 4 vs. 63 +/- 3% with hypoxia alone at 18 h), whereas treatment with anti-IL-8 had no effect. To determine the role of hypoxia-induced ROS on this response, HUVEC monolayers were incubated with the antioxidants ebselen (50 microM) and N-acetyl-L-cysteine (NAC, 1 mM) before hypoxia. Antioxidants attenuated hypoxia-induced IL-6 secretion (13 +/- 2 pg/ml with ebselen and 19 +/- 3 pg/ml with NAC vs. 140 +/- 15 pg/ml with hypoxia). Ebselen and NAC prevented changes in TEER during hypoxia (94 +/- 2% with ebselen and 90 +/- 6% with NAC vs. 63 +/- 3% with hypoxia at 18 h). N-nitro-L-arginine (500 microM) did not decrease hypoxia-induced changes in dichlorofluorescin fluorescence, IL-6 secretion, or TEER. Thus ROS generated during hypoxia act as signaling elements, regulating secretion of the proinflammatory cytokines that lead to alterations of endothelial permeability.


Assuntos
Endotélio Vascular/metabolismo , Hipóxia/metabolismo , Interleucina-6/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/imunologia , Anticorpos/farmacologia , Antioxidantes/farmacologia , Azóis/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/imunologia , Células Cultivadas , Impedância Elétrica , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Humanos , Peróxido de Hidrogênio/farmacologia , Hipóxia/imunologia , Interleucina-6/imunologia , Isoindóis , Compostos Organosselênicos/farmacologia , Oxidantes/farmacologia , Oxigênio/farmacologia , Veias Umbilicais/citologia
19.
J Mol Cell Cardiol ; 31(10): 1885-95, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10525426

RESUMO

Extract from Scutellaria baicalensis Georgi Attenuates Oxidant Stress in Cardiomyocytes. Journal of Molecular and Cellular Cardiology (1999) 31, 1885-1895. Scutellaria baicalensis Georgi is a Chinese herbal medicine used to treat allergic and inflammatory diseases. The medicinal effects of S. baicalensis root may result, in part, from its constituent flavones reported to have antioxidant properties. Since oxidants play multiple roles in cells, we tested whether S. baicalensis could confer protection in a cardiomyocyte model of ischemia and reperfusion. The intracellular fluorescent probes 2',7'-dichlorofluorescin diacetate (DCFH-DA, sensitive to H(2)O(2) and hydroxyl radicals) and dihydroethidium (DHE, sensitive to superoxide) were used to assess intracellular reactive oxygen species (ROS), and propidium iodide (PI) was used to assess viability in cultured embryonic cardiomyocytes. S. baicalensis extract (SbE) quickly attenuated levels of oxidants generated during transient hypoxia and during exposure to the mitochondrial site III inhibitor antimycin A, as measured by DCFH oxidation or by DHE oxidation. These attenuated oxidant levels were associated with improved survival and function. Cell death after ischemia/reperfusion decreased from 47+/-3 % in untreated to 26+/-2 % in S. baicalensis treated cells (P<0.001). After antimycin A exposure, S. baicalensis decreased cell death from 49+/-6 % in untreated to 23+/-4 % in treated cells. Return of contraction occurred in S. baicalensis-treated cells but was not observed in control cells. Other in vitro studies revealed that baicalein, a major flavone component of SbE can directly scavenge superoxide, hydrogen peroxide, and hydroxyl radicals. Collectively, these findings indicate that SbE and its constituent flavones such as baicalein can attenuate oxidant stress and protect cells from lethal oxidant damage in an ischemia-reperfusion model.


Assuntos
Hipóxia Celular , Flavanonas , Flavonoides/farmacologia , Coração/efeitos dos fármacos , Miocárdio/citologia , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/fisiologia , Animais , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas , Embrião de Galinha , Medicamentos de Ervas Chinesas , Ventrículos do Coração , Microscopia de Vídeo , Raízes de Plantas , Plantas Medicinais
20.
FEBS Lett ; 454(3): 173-6, 1999 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-10431801

RESUMO

A resurgence of interest in mitochondrial physiology has recently developed as a result of new experimental data demonstrating that mitochondria function as important participants in a diverse collection of novel intracellular signaling pathways. Cells depleted of mitochondrial DNA, or rho0 cells, lack critical respiratory chain catalytic subunits that are encoded in the mitochondrial genome. Although rho0 cells contain petit mitochondria, they cannot support normal oxidative phosphorylation and must survive and replicate using ATP derived solely from glycolysis. Without a functional electron transport chain, rho0 cells cannot normally regulate redox potential and their mitochondria appear to be incapable of generating reactive oxygen species. Emerging evidence suggests that these signals are important components in a number of mitochondria-initiated signaling pathways. The present article focuses on how rho0 cells have contributed to an understanding of the role that mitochondria play in distinct physiological pathways involved with apoptosis, glucose-induced insulin secretion, and oxygen sensing.


Assuntos
DNA Mitocondrial/fisiologia , Mitocôndrias/fisiologia , Animais , Apoptose , Humanos , Espécies Reativas de Oxigênio/fisiologia , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...