Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Dig Dis ; 28(6): 756-75, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21525761

RESUMEN

Both acute and chronic alcohol consumption increase reactive oxygen species (ROS) formation and lipid peroxidation, whose products damage hepatic mitochondrial DNA (mtDNA). To test whether manganese superoxide dismutase (MnSOD) overexpression modulates acute and chronic alcohol-induced mtDNA lesions, transgenic MnSOD-overexpressing (TgMnSOD(+++)) mice and wild-type (WT) mice were treated by alcohol, either chronically (7 weeks in drinking water) or acutely (single intragastric dose of 5 g/kg). Acute alcohol administration increased mitochondrial ROS formation, decreased mitochondrial glutathione, depleted and damaged mtDNA, durably increased inducible nitric oxide synthase (NOS) expression, plasma nitrites/nitrates and the nitration of tyrosine residues in complex V proteins and decreased complex V activity in WT mice. These effects were prevented in TgMnSOD(+++) mice. In acutely alcoholized WT mice, mtDNA depletion was prevented by tempol, a superoxide scavenger, L-NAME and 1400W, two NOS inhibitors, or uric acid, a peroxynitrite scavenger. In contrast, chronic alcohol consumption decreased cytosolic glutathione and increased hepatic iron, lipid peroxidation products and respiratory complex I protein carbonyls only in ethanol-treated TgMnSOD(+++) mice but not in WT mice. In chronic ethanol-fed TgMnSOD(+++) mice, but not WT mice, mtDNA was damaged and depleted, and the iron chelator, deferoxamine (DFO), prevented this effect. In conclusion, MnSOD overexpression prevents mtDNA depletion after an acute alcohol binge but aggravates this effect after prolonged alcohol consumption, which selectively triggers iron accumulation in TgMnSOD(+++) mice but not in WT mice. In the model of acute alcohol binge, the protective effects of MnSOD, tempol, NOS inhibitors and uric acid suggested a role of the superoxide anion reacting with NO to form mtDNA-damaging peroxynitrite. In the model of prolonged ethanol consumption, the protective effects of DFO suggested the role of iron reacting with hydrogen peroxide to form mtDNA-damaging hydroxyl radical.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , ADN Mitocondrial/metabolismo , Hígado/metabolismo , Superóxido Dismutasa/metabolismo , Animales , ADN Mitocondrial/genética , Hígado/enzimología , Hepatopatías/enzimología , Hepatopatías/genética , Ratones , Estrés Oxidativo
2.
FEBS Lett ; 580(11): 2547-52, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16631755

RESUMEN

Doxorubicin, cis-diamminedichloroplatinum (II) and 5-fluorouracil used in chemotherapy induce apoptosis in Hep3B cells in the absence of p53, p73, and functional Fas. Since mediators remain unknown, the requirement of PKC delta (PKCdelta) and c-Abl was investigated. Suppression of c-Abl or PKCdelta expression using SiRNAs impaired PARP cleavage, Gleevec and/or rottlerin inhibited the induction of the subG1 phase and the increase of reactive oxygen species level. Co-precipitations and phosphorylations to mitochondria of c-Abl, PKCdelta and Bcl-X(L/s) were induced. A depolarization of the mitochondrial membrane and activations of caspase-2 and -9 were observed. We propose that, in the absence of p53, p73 and Fas, genotoxic drugs could require both PKCdelta and c-Abl to induce apoptosis through the mitochondrial pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Fluorouracilo/toxicidad , Mitocondrias/metabolismo , Mutágenos/toxicidad , Proteína Quinasa C-delta/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Caspasa 2 , Caspasa 9 , Caspasas/metabolismo , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cisplatino/toxicidad , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Doxorrubicina/toxicidad , Humanos , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Proteína Quinasa C-delta/genética , Proteínas Proto-Oncogénicas c-abl/genética , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo , Proteína bcl-X/metabolismo , Receptor fas/metabolismo
3.
PLoS One ; 7(7): e40879, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22911714

RESUMEN

During chronic liver inflammation, up-regulated Tumor Necrosis Factor alpha (TNF-α) targets hepatocytes and induces abnormal reactive oxygen species (ROS) production responsible for mitochondrial DNA (mtDNA) alterations. The serine/threonine Glycogen Synthase Kinase 3 beta (GSK3ß) plays a pivotal role during inflammation but its involvement in the maintenance of mtDNA remains unknown. The aim of this study was to investigate its involvement in TNF-α induced mtDNA depletion and its interrelationship with p53 a protein known to maintain mtDNA copy numbers. Using quantitative polymerase chain reaction (qPCR) we found that at 30 min in human hepatoma HepG2 cells TNF-α induced 0.55±0.10 mtDNA lesions per 10 Kb and a 52.4±2.8% decrease in mtDNA content dependent on TNF-R1 receptor and ROS production. Both lesions and depletion returned to baseline from 1 to 6 h after TNF-α exposure. Luminol-amplified chemiluminescence (LAC) was used to measure the rapid (10 min) and transient TNF-α induced increase in ROS production (168±15%). A transient 8-oxo-dG level of 1.4±0.3 ng/mg DNA and repair of abasic sites were also measured by ELISA assays. Translocation of p53 to mitochondria was observed by Western Blot and co-immunoprecipitations showed that TNF-α induced p53 binding to GSK3ß and mitochondrial transcription factor A (TFAM). In addition, mitochondrial D-loop immunoprecipitation (mtDIP) revealed that TNF-α induced p53 binding to the regulatory D-loop region of mtDNA. The knockdown of p53 by siRNAs, inhibition by the phosphoSer(15)p53 antibody or transfection of human mutant active GSK3ßS9A pcDNA3 plasmid inhibited recovery of mtDNA content while blockade of GSK3ß activity by SB216763 inhibitor or knockdown by siRNAs suppressed mtDNA depletion. This study is the first to report the involvement of GSK3ß in TNF-α induced mtDNA depletion. We suggest that p53 binding to GSK3ß, TFAM and D-loop could induce recovery of mtDNA content through mtDNA repair.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , ADN Mitocondrial , Glucógeno Sintasa Quinasa 3/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Apoptosis/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células Hep G2 , Humanos , Proteínas Mitocondriales/metabolismo , Unión Proteica , Transporte de Proteínas , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
4.
Antioxid Redox Signal ; 15(11): 2837-54, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21767162

RESUMEN

Hepatic energy depletion has been described in severe sepsis, and lipopolysaccharide (LPS) has been shown to cause mitochondrial DNA (mtDNA) damage. To clarify the mechanisms of LPS-induced mtDNA damage and mitochondrial alterations, we treated wild-type (WT) or transgenic manganese superoxide dismutase-overerexpressing (MnSOD(+++)) mice with a single dose of LPS (5 mg/kg). In WT mice, LPS increased mitochondrial reactive oxygen species formation, hepatic inducible nitric oxide synthase (NOS) mRNA and protein, tumor necrosis factor-alpha, interleukin-1 beta, and high-mobility group protein B1 concentrations. Six to 48 h after LPS administration (5 mg/kg), liver mtDNA levels, respiratory complex I activity, and adenosine triphosphate (ATP) contents were decreased. In addition, LPS increased interferon-ß concentration and decreased mitochondrial transcription factor A (Tfam) mRNA, Tfam protein, and mtDNA-encoded mRNAs. Morphological studies showed mild hepatic inflammation. The LPS (5 mg/kg)-induced mtDNA depletion, complex I inactivation, ATP depletion, and alanine aminotransferase increase were prevented in MnSOD(+++) mice or in WT mice cotreated with 1400W (a NOS inhibitor), (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride, monohydrate (a superoxide scavenger) or uric acid (a peroxynitrite scavenger). The MnSOD overexpression delayed death in mice challenged by a higher, lethal dose of LPS (25 mg/kg). In conclusion, LPS administration damages mtDNA and alters mitochondrial function. The protective effects of MnSOD, NOS inhibitors, and superoxide or peroxynitrite scavengers point out a role of the superoxide anion reacting with NO to form mtDNA- and protein-damaging peroxynitrite. In addition to the acute damage caused by reactive species, decreased levels of mitochondrial transcripts contribute to mitochondrial dysfunction.


Asunto(s)
ADN Mitocondrial/metabolismo , Lipopolisacáridos/farmacología , Complejos de ATP Sintetasa/metabolismo , Aconitato Hidratasa/metabolismo , Adenosina Trifosfato/metabolismo , Alanina Transaminasa/sangre , Animales , Proteínas de Unión al ADN/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Células Hep G2 , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Interferón beta/sangre , Interferón beta/farmacología , Hierro/sangre , Hierro/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nitratos/sangre , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/sangre , Especies Reactivas de Oxígeno/metabolismo , Sepsis/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Receptor Toll-Like 4/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Factor de Necrosis Tumoral alfa/sangre , Tirosina/análogos & derivados , Tirosina/metabolismo
5.
J Hepatol ; 42(2): 230-7, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15664249

RESUMEN

BACKGROUND/AIMS: The efficacy of a targeted cytotoxic hybrid somatostatin analogue AN-238 and of its superactive radical 2-pyrrolinodoxorubicin (AN-201) to induce apoptosis of HepG2 and Hep3B human hepatoma cell lines were studied. AN-238 was designed to selectively target tumor cells expressing somatostatin receptor subtypes (sst(s)). Its effects on HepG2 or Hep3B cells displaying or lacking tumor suppressor p53, respectively, were compared. Normal rat isolated hepatocytes were also tested. METHODS: sst(s) were characterized by binding assays and RT-PCR. Cytotoxicity was quantified by flow cytometry. DNA fragmentation was studied by gel electrophoresis, PARP cleavage by Western blot and ROS formation using fluorescent probes. RESULTS: Specific binding of iodinated RC-160 to HepG2 and Hep3B cells, and its displacement by AN-238 was characterized. mRNA for hsst(2A) was found in both cell lines. Flow cytometry showed a stronger effect of AN-238 than AN-201 to induce sub-G1 phase. DNA fragmentation, nuclear bodies, and PARP cleavage were observed. In addition, AN-238 increased formation of ROS more potently than AN-201. However, no inductions of DNA fragmentation by AN-201 or AN-238 were observed on rat hepatocytes. CONCLUSIONS: Our results indicate that, in liver cancer, the cytotoxic somatostatin analogue AN-238 is a powerful agent that can induce apoptosis, through sst(s) and independently of p53.


Asunto(s)
Apoptosis/efectos de los fármacos , Citotoxinas/toxicidad , Doxorrubicina/toxicidad , Pirroles/toxicidad , Receptores de Somatostatina/antagonistas & inhibidores , Secuencia de Bases , Carcinoma Hepatocelular , Línea Celular Tumoral , Cartilla de ADN , Doxorrubicina/análogos & derivados , Humanos , Neoplasias Hepáticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Somatostatina/genética
6.
Biochem J ; 364(Pt 3): 881-5, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12049655

RESUMEN

The chemotherapeutic drug doxorubicin and the anti-proliferative long-acting somatostatin analogue octreotide, both used in cancer treatment, have been shown to increase the expression of the p53 tumour suppressor protein. In the present study, we demonstrate by Western-blot analysis that, in addition to the p53 protein, these molecules were able to induce the expression of a shorter protein with an apparent molecular mass of 40 kDa (p40), recognized by antibodies raised against the N-terminus of p53. This induction was present in tumoral and non-tumoral cells and did not depend on the status of the endogenous p53 protein. The p40 protein was significantly induced after 3 h of cell treatment with doxorubicin or octreotide, remained stable until 24 h and was located in the nuclear extract. Using reverse primers corresponding to each exon of the p53 gene, only one transcript was amplified by reverse transcriptase-PCR. This suggested that p40 was issued from a post-translational modification and not from an alternative splicing. This protein was not recognized by the PAb421 antibody, suggesting that it was issued from a cleavage of the p53 C-terminal region (p40deltaC). Furthermore, this cleavage was not dependent on caspase activity. In conclusion, these results support the hypothesis that this post-translational modification plays a significant role in the regulation of multiple p53 signalling pathways. These results also suggest that octreotide, a molecule with different signalling pathways, was able as doxorubicin to generate a p53 breakdown product.


Asunto(s)
Doxorrubicina/farmacología , Genes p53/efectos de los fármacos , Octreótido/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Carcinoma Hepatocelular , Neoplasias del Colon , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Hepáticas , Peso Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda