Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
FASEB J ; 25(8): 2669-87, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21551238

RESUMEN

Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) is a mitochondrial disease most usually caused by point mutations in tRNA genes encoded by mtDNA. Here, we report on how this mutation affects mitochondrial function in primary fibroblast cultures established from 2 patients with MELAS who harbored the A3243G mutation. Both mitochondrial respiratory chain enzyme activities and coenzyme Q(10) (CoQ) levels were significantly decreased in MELAS fibroblasts. A similar decrease in mitochondrial membrane potential was found in intact MELAS fibroblasts. Mitochondrial dysfunction was associated with increased oxidative stress and the activation of mitochondrial permeability transition (MPT), which triggered the degradation of impaired mitochondria. Furthermore, we found defective autophagosome elimination in MELAS fibroblasts. Electron and fluorescence microscopy studies confirmed a massive degradation of mitochondria and accumulation of autophagosomes, suggesting mitophagy activation and deficient autophagic flux. Transmitochondrial cybrids harboring the A3243G mutation also showed CoQ deficiency and increased autophagy activity. All these abnormalities were partially restored by CoQ supplementation. Autophagy in MELAS fibroblasts was also abolished by treatment with antioxidants or cyclosporine, suggesting that both reactive oxygen species and MPT participate in this process. Furthermore, prevention of autophagy in MELAS fibroblasts resulted in apoptotic cell death, suggesting a protective role of autophagy in MELAS fibroblasts.


Asunto(s)
Síndrome MELAS/metabolismo , Síndrome MELAS/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Ubiquinona/análogos & derivados , Autofagia/genética , Autofagia/fisiología , Proteína 5 Relacionada con la Autofagia , Secuencia de Bases , Células Cultivadas , Cartilla de ADN/genética , ADN Mitocondrial/genética , Transporte de Electrón , Fibroblastos/metabolismo , Fibroblastos/patología , Técnicas de Silenciamiento del Gen , Humanos , Síndrome MELAS/genética , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Mutación Puntual , ARN Interferente Pequeño/genética , ARN de Transferencia de Leucina/genética , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/deficiencia
2.
Apoptosis ; 16(4): 404-24, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21311976

RESUMEN

Microtubule cytoskeleton is reformed during apoptosis, forming a cortical structure beneath plasma membrane, which plays an important role in preserving cell morphology and plasma membrane integrity. However, the maintenance of the apoptotic microtubule network (AMN) during apoptosis is not understood. In the present study, we examined apoptosis induced by camptothecin (CPT), a topoisomerase I inhibitor, in human H460 and porcine LLCPK-1α cells. We demonstrate that AMN was organized in apoptotic cells with high ATP levels and hyperpolarized mitochondria and, on the contrary, was dismantled in apoptotic cells with low ATP levels and mitochondrial depolarization. AMN disorganization after mitochondrial depolarization was associated with increased plasma membrane permeability assessed by enhancing LDH release and increased intracellular calcium levels. Living cell imaging monitoring of both, microtubule dynamics and mitochondrial membrane potential, showed that AMN persists during apoptosis coinciding with cycles of mitochondrial hyperpolarization. Eventually, AMN was disorganized when mitochondria suffered a large depolarization and cell underwent secondary necrosis. AMN stabilization by taxol prevented LDH release and calcium influx even though mitochondria were depolarized, suggesting that AMN is essential for plasma membrane integrity. Furthermore, high ATP levels and mitochondria polarization collapse after oligomycin treatment in apoptotic cells suggest that ATP synthase works in "reverse" mode during apoptosis. These data provide new explanations for the role of AMN and mitochondria during apoptosis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Apoptosis , Metabolismo Energético , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Actinas/metabolismo , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Línea Celular Tumoral , Colchicina/farmacología , Citocromos c/metabolismo , Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos , Cinética , L-Lactato Deshidrogenasa/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Oligomicinas/farmacología , ATPasas de Translocación de Protón/metabolismo
3.
Toxicol Appl Pharmacol ; 235(3): 329-37, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19263520

RESUMEN

Since amitriptyline is a very frequently prescribed antidepressant drug, it is not surprising that amitriptyline toxicity is relatively common. Amitriptyline toxic systemic effects include cardiovascular, autonomous nervous, and central nervous systems. To understand the mechanisms of amitriptyline toxicity we studied the cytotoxic effects of amitriptyline treatment on cultured primary human fibroblasts and zebrafish embryos, and the protective role of coenzyme Q(10) and alpha-tocopherol, two membrane antioxidants. We found that amitriptyline treatment induced oxidative stress and mitochondrial dysfunction in primary human fibroblasts. Mitochondrial dysfunction in amitriptyline treatment was characterized by reduced expression levels of mitochondrial proteins and coenzyme Q(10), decreased NADH:cytochrome c reductase activity, and a drop in mitochondrial membrane potential. Moreover, and as a consequence of these toxic effects, amitriptyline treatment induced a significant increase in apoptotic cell death activating mitochondrial permeability transition. Coenzyme Q(10) and alpha-tocopherol supplementation attenuated ROS production, lipid peroxidation, mitochondrial dysfunction, and cell death, suggesting that oxidative stress affecting cell membrane components is involved in amitriptyline cytotoxicity. Furthermore, amitriptyline-dependent toxicity and antioxidant protection were also evaluated in zebrafish embryos, a well established vertebrate model to study developmental toxicity. Amitriptyline significantly increased embryonic cell death and apoptosis rate, and both antioxidants provided a significant protection against amitriptyline embryotoxicity.


Asunto(s)
Amitriptilina/antagonistas & inhibidores , Amitriptilina/toxicidad , Ubiquinona/análogos & derivados , alfa-Tocoferol/farmacología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/farmacología , Pez Cebra/embriología
4.
Free Radic Biol Med ; 40(8): 1293-302, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16631519

RESUMEN

Free radicals have been implicated in the action of many chemotherapeutic drugs. Here we tested the hypothesis that camptothecin and other chemotherapeutic drugs, such as etoposide, doxorubicin, and methotrexate, induce an increase in coenzyme Q(10) levels as part of the antioxidant defense against free radical production under these anticancer treatments in cancer cell lines. Chemotherapy treatment induced both free radical production and an increase in coenzyme Q(10) levels in all the cancer cell lines tested. Reduced coenzyme Q(10) form levels were particularly enhanced. Coenzyme Q(10)-increased levels were associated with up-regulation of COQ genes expression, involved in coenzyme Q(10) biosynthesis. At the translational level, COQ7 protein expression levels were also increased. Furthermore, coenzyme Q(10) biosynthesis inhibition blocked camptothecin-induced coenzyme Q(10) increase, and enhanced camptothecin cytotoxicity. Our findings suggest that coenzyme Q(10) increase is implicated in the cellular defense under chemotherapy treatment and may contribute to cell survival.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/farmacología , Ubiquinona/análogos & derivados , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Camptotecina/toxicidad , Línea Celular Tumoral , Coenzimas , Radicales Libres/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Oxidación-Reducción/efectos de los fármacos , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/biosíntesis , Regulación hacia Arriba/efectos de los fármacos
5.
Redox Biol ; 5: 420-421, 2015 08.
Artículo en Inglés | MEDLINE | ID: mdl-28162288

RESUMEN

Hepatocellular carcinoma develops in cirrhotic liver. The nitric oxide (NO) synthase type III (NOS-3) overexpression induces cell death in hepatoma cells. The study developed gene therapy designed to specifically overexpress NOS-3 in cultured hepatoma cells, and in tumors derived from orthotopically implanted tumor cells in fibrotic livers. Liver fibrosis was induced by CCl4 administration in mice. Hepa 1-6 cells were used for in vitro and in vivo experiments. The first generation adenovirus was designed to overexpress NOS-3 (or GFP) and luciferase cDNA under the regulation of murine alpha-fetoprotein (AFP) and Rous Sarcoma Virus (RSV) promoters, respectively. Both adenoviruses were administered through the tail vein two weeks after orthotopic tumor cell implantation. AFP-NOS-3/RSV-Luciferase increased oxidative-related DNA damage, p53, CD95/CD95L expression and caspase-8 activity in cultured Hepa 1-6 cells. The increased expression of CD95/CD95L and caspase-8 activity was abolished by l-NAME or p53 siRNA. The tail vein infusion of AFP-NOS- 3/RSV-Luciferase adenovirus increased cell death markers, and reduced cell proliferation of established tumors in fibrotic livers. The increase of oxidative/nitrosative stress induced by NOS-3 overexpression induced DNA damage, p53, CD95/CD95L expression and cell death in hepatocellular carcinoma cells. The effectiveness of the gene therapy has been demonstrated in vitro and in vivo.


Asunto(s)
Adenoviridae , Carcinoma Hepatocelular/terapia , Terapia Genética , Neoplasias Hepáticas Experimentales/terapia , Óxido Nítrico Sintasa de Tipo III , Animales , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas Experimentales/enzimología , Neoplasias Hepáticas Experimentales/genética , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/genética
6.
Autophagy ; 5(1): 19-32, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19115482

RESUMEN

Coenzyme Q10 (CoQ) is a small lipophilic molecule critical for the transport of electrons from complexes I and II to complex III in the mitochondrial respiratory chain. CoQ deficiency is a rare human genetic condition that has been associated with a variety of clinical phenotypes. With the aim of elucidating how CoQ deficiency affects an organism, we have investigated the pathophysiologic processes present within fibroblasts derived from 4 patients with CoQ deficiency. Assays of cultured fibroblasts revealed decreased activities of complex II+III, complex III, and complex IV, reduced expression of mitochondrial proteins involved in oxidative phosphorylation, decreased mitochondrial membrane potential, increased production of reactive oxygen species (ROS), activation of mitochondrial permeability transition (MPT), and reduced growth rates. These abnormalities were partially restored by CoQ supplementation. Moreover, we demonstrate that CoQ deficient fibroblasts exhibited increased levels of lysosomal markers (beta-galactosidase, cathepsin, LC3, and Lyso Tracker), and enhanced expression of autophagic genes at both transcriptional and translational levels, indicating the presence of autophagy. Electron microscopy studies confirmed a massive degradation of the altered mitochondria by mitophagy. Autophagy in CoQ deficient fibroblasts was abolished by antioxidants or cyclosporin treatments suggesting that both ROS and MPT participate in this process. Furthermore, prevention of autophagy in CoQ deficient fibroblasts by 3-methyl adenine or wortmannin, as well as the induction of CoQ deficiency in cells lacking autophagy (by means of genetic knockout of the Atg5 gene in mouse embryonic fibroblasts) resulted in apoptotic cell death, suggesting a protective role of autophagy in CoQ deficiency.


Asunto(s)
Autofagia , Mitocondrias/metabolismo , Ubiquinona/deficiencia , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Biomarcadores/metabolismo , Western Blotting , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibroblastos/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/ultraestructura , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Proteínas Mitocondriales/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Síndrome , Ubiquinona/farmacología
7.
Apoptosis ; 12(7): 1195-208, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17245640

RESUMEN

It has recently been shown that the microtubule cytoskeleton is reformed during the execution phase of apoptosis. We demonstrate that this microtubule reformation occurs in many cell types and under different apoptotic stimuli. We confirm that the apoptotic microtubule network possesses a novel organization, whose nucleation appears independent of conventional gamma-tubulin ring complex containing structures. Our analysis suggests that microtubules are closely associated with the plasma membrane, forming a cortical ring or cellular "cocoon". Concomitantly other components of the cytoskeleton, such as actin and cytokeratins disassemble. We found that colchicine-mediated disruption of apoptotic microtubule network results in enhanced plasma membrane permeability and secondary necrosis, suggesting that the reformation of a microtubule cytoskeleton plays an important role in preserving plasma membrane integrity during apoptosis. Significantly, cells induced to enter apoptosis in the presence of the pan-caspase inhibitor z-VAD, nevertheless form microtubule-like structures suggesting that microtubule formation is not dependent on caspase activation. In contrast we found that treatment with EGTA-AM, an intracellular calcium chelator, prevents apoptotic microtubule network formation, suggesting that intracellular calcium may play an essential role in the microtubule reformation. We propose that apoptotic microtubule network is required to maintain plasma membrane integrity during the execution phase of apoptosis.


Asunto(s)
Apoptosis/fisiología , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Actinas/metabolismo , Calcio/metabolismo , Caspasas/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Colchicina/farmacología , Citoesqueleto/metabolismo , Humanos , Filamentos Intermedios/metabolismo , Tubulina (Proteína)/metabolismo
8.
Hum Mol Genet ; 16(9): 1091-7, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17374725

RESUMEN

Coenzyme Q(10) (CoQ(10)) deficiency has been associated with an increasing number of clinical phenotypes that respond to CoQ(10) supplementation. In two siblings with encephalomyopathy, nephropathy and severe CoQ(10) deficiency, a homozygous mutation was identified in the CoQ(10) biosynthesis gene COQ2, encoding polyprenyl-pHB transferase. To confirm the pathogenicity of this mutation, we have demonstrated that human wild-type, but not mutant COQ2, functionally complements COQ2 defective yeast. In addition, an equivalent mutation introduced in the yeast COQ2 gene also decreases both CoQ(6) concentration and growth in respiratory-chain dependent medium. Polyprenyl-pHB transferase activity was 33-45% of controls in COQ2 mutant fibroblasts. CoQ-dependent mitochondrial complexes activities were restored in deficient fibroblasts by CoQ(10) supplementation, and growth rate was restored in these cells by either CoQ(10) or uridine supplementation. This work is the first direct demonstration of the pathogenicity of a COQ2 mutation involved in human disease, and establishes yeast as a useful model to study human CoQ(10) deficiency. Moreover, we demonstrate that CoQ(10) deficiency in addition to the bioenergetics defect also impairs de novo pyrimidine synthesis, which may contribute to the pathogenesis of the disease.


Asunto(s)
Transferasas Alquil y Aril/genética , Metabolismo Energético/genética , Mutación Missense , Pirimidinas/biosíntesis , Transferasas Alquil y Aril/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , División Celular/efectos de los fármacos , División Celular/genética , Células Cultivadas , Coenzimas/biosíntesis , Coenzimas/metabolismo , Coenzimas/farmacología , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Prueba de Complementación Genética , Células HeLa , Humanos , Immunoblotting , Mitocondrias/metabolismo , Datos de Secuencia Molecular , Prohibitinas , Pirimidinas/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Alineación de Secuencia , Ubiquinona/análogos & derivados , Ubiquinona/biosíntesis , Ubiquinona/metabolismo , Ubiquinona/farmacología , Uridina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA