Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Lab Invest ; 92(3): 396-410, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22157718

RESUMO

Although carbon tetrachloride (CCl(4))-induced acute and chronic hepatotoxicity have been extensively studied, little is known about the very early in vivo effects of this organic solvent on oxidative stress and mitochondrial function. In this study, mice were treated with CCl(4) (1.5 ml/kg ie 2.38 g/kg) and parameters related to liver damage, lipid peroxidation, stress/defense and mitochondria were studied 3 h later. Some CCl(4)-intoxicated mice were also pretreated with the cytochrome P450 2E1 inhibitor diethyldithiocarbamate or the antioxidants Trolox C and dehydroepiandrosterone. CCl(4) induced a moderate elevation of aminotransferases, swelling of centrilobular hepatocytes, lipid peroxidation, reduction of cytochrome P4502E1 mRNA levels and a massive increase in mRNA expression of heme oxygenase-1 and heat shock protein 70. Moreover, CCl(4) intoxication induced a severe decrease of mitochondrial respiratory chain complex IV activity, mitochondrial DNA depletion and damage as well as ultrastructural alterations. Whereas DDTC totally or partially prevented all these hepatic toxic events, both antioxidants protected only against liver lipid peroxidation and mitochondrial damage. Taken together, our results suggest that lipid peroxidation is primarily implicated in CCl(4)-induced early mitochondrial injury. However, lipid peroxidation-independent mechanisms seem to be involved in CCl(4)-induced early hepatocyte swelling and changes in expression of stress/defense-related genes. Antioxidant therapy may not be an efficient strategy to block early liver damage after CCl(4) intoxication.


Assuntos
Intoxicação por Tetracloreto de Carbono/metabolismo , Hepatócitos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Animais , Antioxidantes/farmacologia , Tetracloreto de Carbono , Cromanos/farmacologia , Inibidores do Citocromo P-450 CYP2E1 , Desidroepiandrosterona/farmacologia , Ditiocarb/farmacologia , Masculino , Camundongos , Mitocôndrias Hepáticas/efeitos dos fármacos
2.
Drug Metab Rev ; 44(1): 34-87, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21892896

RESUMO

A frequent mechanism for drug-induced liver injury (DILI) is the formation of reactive metabolites that trigger hepatitis through direct toxicity or immune reactions. Both events cause mitochondrial membrane disruption. Genetic or acquired factors predispose to metabolite-mediated hepatitis by increasing the formation of the reactive metabolite, decreasing its detoxification, or by the presence of critical human leukocyte antigen molecule(s). In other instances, the parent drug itself triggers mitochondrial membrane disruption or inhibits mitochondrial function through different mechanisms. Drugs can sequester coenzyme A or can inhibit mitochondrial ß-oxidation enzymes, the transfer of electrons along the respiratory chain, or adenosine triphosphate (ATP) synthase. Drugs can also destroy mitochondrial DNA, inhibit its replication, decrease mitochondrial transcripts, or hamper mitochondrial protein synthesis. Quite often, a single drug has many different effects on mitochondrial function. A severe impairment of oxidative phosphorylation decreases hepatic ATP, leading to cell dysfunction or necrosis; it can also secondarily inhibit ß-oxidation, thus causing steatosis, and can also inhibit pyruvate catabolism, leading to lactic acidosis. A severe impairment of ß-oxidation can cause a fatty liver; further, decreased gluconeogenesis and increased utilization of glucose to compensate for the inability to oxidize fatty acids, together with the mitochondrial toxicity of accumulated free fatty acids and lipid peroxidation products, may impair energy production, possibly leading to coma and death. Susceptibility to parent drug-mediated mitochondrial dysfunction can be increased by factors impairing the removal of the toxic parent compound or by the presence of other medical condition(s) impairing mitochondrial function. New drug molecules should be screened for possible mitochondrial effects.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , Respiração Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/patologia , DNA Mitocondrial/biossíntese , DNA Mitocondrial/efeitos dos fármacos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Humanos , Sistema Imunitário/metabolismo , Fígado/patologia , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/patologia , Mitocôndrias Hepáticas/ultraestrutura , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/efeitos dos fármacos , Oxirredução , Preparações Farmacêuticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
3.
J Pharmacol Exp Ther ; 332(3): 886-97, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20016022

RESUMO

Alcohol consumption increases reactive oxygen species (ROS) formation, which can damage mitochondrial DNA (mtDNA) and alter mitochondrial function. To test whether manganese superoxide dismutase (MnSOD) modulates acute alcohol-induced mitochondrial alterations, transgenic MnSOD-overexpressing (MnSOD(+++)) mice, heterozygous knockout (MnSOD(+/-)) mice, and wild-type (WT) littermates were sacrificed 2 or 24 h after intragastric ethanol administration (5 g/kg). Alcohol administration further increased MnSOD activity in MnSOD(+++) mice, but further decreased it in MnSOD(+/-) mice. In WT mice, alcohol administration transiently increased mitochondrial ROS formation, decreased mitochondrial glutathione, depleted and damaged mtDNA, and decreased complex I and V activities; alcohol durably increased inducible nitric-oxide synthase (NOS) expression, plasma nitrites/nitrates, and the nitration of tyrosine residues in complex V proteins. These effects were prevented in MnSOD(+++) mice and prolonged in MnSOD(+/-) mice. In alcoholized WT or MnSOD(+/-) mice, mtDNA depletion and the nitration of tyrosine residues in complex I and V proteins were prevented or attenuated by cotreatment with tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl), a superoxide scavenger; N(omega)-nitro-l-arginine methyl ester and N-[3-(aminomethyl)benzyl]acetamidine (1,400W), two NOS inhibitors; or uric acid, a peroxynitrite scavenger. In conclusion, MnSOD overexpression prevents, and MnSOD deficiency prolongs, mtDNA depletion after an acute alcohol binge in mice. The protective effects of MnSOD, tempol, NOS inhibitors, and uric acid point out a role of the superoxide anion reacting with NO to form mtDNA-damaging peroxynitrite.


Assuntos
DNA Mitocondrial/metabolismo , Etanol/intoxicação , Fígado/metabolismo , Ácido Peroxinitroso/metabolismo , Superóxido Dismutase/fisiologia , Animais , Caspase 3/metabolismo , Óxidos N-Cíclicos/farmacologia , Proteínas de Ligação a DNA/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Sequestradores de Radicais Livres/farmacologia , Glutationa Peroxidase/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Ferro/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/fisiologia , ATPases Mitocondriais Próton-Translocadoras/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Marcadores de Spin , Superóxido Dismutase/biossíntese , Fatores de Transcrição/metabolismo , Ácido Úrico/farmacologia
4.
Handb Exp Pharmacol ; (196): 311-65, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20020267

RESUMO

Mitochondrial dysfunction is a major mechanism of liver injury. A parent drug or its reactive metabolite can trigger outer mitochondrial membrane permeabilization or rupture due to mitochondrial permeability transition. The latter can severely deplete ATP and cause liver cell necrosis, or it can instead lead to apoptosis by releasing cytochrome c, which activates caspases in the cytosol. Necrosis and apoptosis can trigger cytolytic hepatitis resulting in lethal fulminant hepatitis in some patients. Other drugs severely inhibit mitochondrial function and trigger extensive microvesicular steatosis, hypoglycaemia, coma, and death. Milder and more prolonged forms of drug-induced mitochondrial dysfunction can also cause macrovacuolar steatosis. Although this is a benign liver lesion in the short-term, it can progress to steatohepatitis and then to cirrhosis. Patient susceptibility to drug-induced mitochondrial dysfunction and liver injury can sometimes be explained by genetic or acquired variations in drug metabolism and/or elimination that increase the concentration of the toxic species (parent drug or metabolite). Susceptibility may also be increased by the presence of another condition, which also impairs mitochondrial function, such as an inborn mitochondrial cytopathy, beta-oxidation defect, certain viral infections, pregnancy, or the obesity-associated metabolic syndrome. Liver injury due to mitochondrial dysfunction can have important consequences for pharmaceutical companies. It has led to the interruption of clinical trials, the recall of several drugs after marketing, or the introduction of severe black box warnings by drug agencies. Pharmaceutical companies should systematically investigate mitochondrial effects during lead selection or preclinical safety studies.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Mitocôndrias Hepáticas/efeitos dos fármacos , Doenças Mitocondriais/induzido quimicamente , Animais , Apoptose/efeitos dos fármacos , Biotransformação , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Dano ao DNA , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Metabolismo Energético/efeitos dos fármacos , Humanos , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/patologia , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Proteínas de Transporte da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Necrose , Estresse Oxidativo/efeitos dos fármacos
5.
Toxicol Appl Pharmacol ; 231(3): 336-43, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18572215

RESUMO

Fas stimulation recruits neutrophils and activates macrophages that secrete tumor necrosis factor-alpha (TNF-alpha), which aggravates Fas-mediated liver injury. To determine whether nonsteroidal anti-inflammatory drugs modify these processes, we challenged 24-hour-fasted mice with the agonistic Jo2 anti-Fas antibody (4 microg/mouse), and treated the animals 1 h later with saline or ibuprofen (250 mg/kg), a dual cyclooxygenase (COX)-1 and COX-2 inhibitor. Ibuprofen attenuated the Jo2-mediated recruitment/activation of myeloperoxidase-secreting neutrophils/macrophages in the liver, and attenuated the surge in serum TNF-alpha. Ibuprofen also minimized hepatic glutathione depletion, Bid truncation, caspase activation, outer mitochondrial membrane rupture, hepatocyte apoptosis and the increase in serum alanine aminotransferase (ALT) activity 5 h after Jo2 administration, to finally decrease mouse mortality at later times. The concomitant administration of pentoxifylline (decreasing TNF-alpha secretion) and infliximab (trapping TNF-alpha) likewise attenuated the Jo2-mediated increase in TNF-alpha, the decrease in hepatic glutathione, and the increase in serum ALT activity 5 h after Jo2 administration. The concomitant administration of the COX-1 inhibitor, SC-560 (10 mg/kg) and the COX-2 inhibitor, celecoxib (40 mg/kg) 1 h after Jo2 administration, also decreased liver injury 5 h after Jo2 administration. In contrast, SC-560 (10 mg/kg) or celecoxib (40 or 160 mg/kg) given alone had no significant protective effects. In conclusion, secondary TNF-alpha secretion plays an important role in Jo2-mediated glutathione depletion and liver injury. The combined inhibition of COX-1 and COX-2 by ibuprofen attenuates TNF-alpha secretion, glutathione depletion, mitochondrial alterations, hepatic apoptosis and mortality in Jo2-treated fasted mice.


Assuntos
Apoptose/fisiologia , Glutationa/deficiência , Hepatite/metabolismo , Ibuprofeno/administração & dosagem , Fator de Necrose Tumoral alfa/sangue , Receptor fas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Hepatite/enzimologia , Hepatite/mortalidade , Hepatite/patologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Ibuprofeno/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
6.
Toxicol In Vitro ; 22(6): 1511-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18603402

RESUMO

Mallory-Denk bodies (MDB) are hepatocyte inclusions containing cytokeratin 8 (CK8) which can develop, along with other steatohepatitis lesions, in patients treated with amiodarone, perhexiline maleate or 4,4'-diethylaminoethoxyhexestrol. These drugs accumulate lipids, whose subsequent peroxidation liberates reactive by-products, like malondialdehyde (MDA). The formation of MDB has been previously reproduced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine or griseofulvin administration which cross-link CK8 by tissue transglutaminase, thus forming an entangled network, from which MDB progressively arise. The present study depicts the mechanisms initiating MDB formation by steatohepatitis-inducing drugs. Short incubation of hepatocytes with amiodarone (50 microM), 4,4'-diethylaminoethoxyhexestrol (50 microM) or perhexiline maleate (25 microM) increased the pool of CK8 monomers and increased cell calcium to activate Ca(++)-dependent transglutaminases which cross-linked the CK8 monomers into CK8-containing oligomers. The present study also provides the first evidence that MDA might directly participate in MDB formation, as this reactive agent cross-linked purified CK8 or albumin in vitro, disrupted the cytokeratin network of isolated hepatocytes, and bridged CK8 molecules. In conclusion, steatohepatitis-inducing drugs increase cell calcium and activate tissue transglutaminase, which cross-links CK8 to form a molecular scaffold, from which MDB might secondarily arise. Malondialdehyde also cross-links CK8, albeit through a different mechanism, and might also contribute to MDB formation.


Assuntos
Hepatócitos/efeitos dos fármacos , Corpos de Inclusão/efeitos dos fármacos , Queratina-8/efeitos dos fármacos , Malondialdeído/metabolismo , Amiodarona/toxicidade , Animais , Cálcio/metabolismo , Fígado Gorduroso/induzido quimicamente , Proteínas de Ligação ao GTP/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Hepatócitos/metabolismo , Hexestrol/análogos & derivados , Hexestrol/toxicidade , Corpos de Inclusão/metabolismo , Queratina-8/metabolismo , Masculino , Perexilina/análogos & derivados , Perexilina/toxicidade , Proteína 2 Glutamina gama-Glutamiltransferase , Proteínas , Ratos , Ratos Sprague-Dawley , Transglutaminases/efeitos dos fármacos , Transglutaminases/metabolismo
7.
Toxicol In Vitro ; 22(3): 730-46, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18191936

RESUMO

OBJECTIVE: To investigate the possible mechanisms underlying the liver enzyme elevations seen during clinical studies of long-term treatment (>35 days) with ximelagatran, and investigate the usefulness of pre-clinical in vitro systems to predict drug-induced liver effects. METHODS: Ximelagatran and its metabolites were tested for effects on cell viability, mitochondrial function, formation of reactive metabolites and reactive oxygen species, protein binding, and induction of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) gene expression or nuclear orphan receptors. Experimental systems included fresh and cryopreserved hepatocytes, human hepatoma cell lines (HepG2 and HuH-7) and subcellular human liver fractions. RESULTS: Loss of cell viability was only seen in HepG2 cells at ximelagatran concentrations 100 microM and in cryopreserved human hepatocytes at 300 microM, while HuH-7 cells were not affected by 24 h exposure at up to 300 microM ximelagatran. Calcium homeostasis was not affected in HepG2 cells exposed to ximelagatran up to 300 microM for 15 min. There was no evidence for the formation of reactive metabolites when cell systems were exposed to ximelagatran. ALT and AST expression in human hepatoma cell lines were also unchanged by ximelagatran. Mitochondrial functions such as respiration, opening of the transition pore, mitochondrial membrane depolarization and beta-oxidation were not affected by ximelagatran or its metabolites. CONCLUSION: Ximelagatran at concentrations considerably higher than that found in plasma following therapeutic dosing had little or no effect on cellular functions studied in vitro. The in vitro studies therefore did not elucidate the mechanism by which ximelagatran induces liver effects in humans, possibly because of limitations in the experimental systems not reflecting characteristics of the human hepatocyte, restricted exposure time, or because the primary mechanism for the observed clinical liver effects is not on the parenchymal liver cell.


Assuntos
Azetidinas/toxicidade , Benzilaminas/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fibrinolíticos/toxicidade , Trombina/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Azetidinas/metabolismo , Benzilaminas/metabolismo , Cálcio/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular , Cromatografia Líquida , Criopreservação , Fibrinolíticos/metabolismo , Citometria de Fluxo , Hepatócitos/efeitos dos fármacos , Humanos , Técnicas In Vitro , Espectrometria de Massas , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Necrose , Permeabilidade , Valor Preditivo dos Testes , Ratos , Espécies Reativas de Oxigênio/metabolismo , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo
8.
J Hepatol ; 46(5): 858-68, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17275124

RESUMO

BACKGROUND/AIMS: The agonistic Jo2 anti-Fas antibody reproduces human fulminant hepatitis in mice. We tested the hypothesis that enhancing hepatic glutathione (GSH) stores may prevent Jo2-induced apoptosis. METHODS: We fed mice with a normal diet or a sulfur amino acid-enriched (SAA(+)) diet increasing hepatic GSH by 63%, and challenged these mice with Jo2. RESULTS: The SAA(+) diet markedly attenuated the Jo2-mediated decrease in hepatic GSH and the increase in the oxidized glutathione (GSSG)/GSH ratio in cytosol and mitochondria. The SAA(+) diet prevented protein kinase Czeta (PKCzeta) and p47(phox) phosphorylations, Yes activation, Fas-tyrosine phosphorylation, Bid truncation, Bax, and cytochrome c translocations, the mitochondrial membrane potential collapse, caspase activation, DNA fragmentation, hepatocyte apoptosis, and mouse lethality after Jo2 administration. The protective effect of the SAA(+) diet was abolished by a small dose of phorone decreasing hepatic GSH back to the levels observed in mice fed the normal diet. Conversely, administration of GSH monoethyl ester after Jo2 administration prevented hepatic GSH depletion and attenuated toxicity in mice fed with the normal diet. CONCLUSIONS: The SAA(+) diet preserves GSSG/GSH ratios, and prevents PKCzeta and p47(phox) phosphorylations, Yes activation, Fas-tyrosine phosphorylation, mitochondrial permeabilization, and hepatic apoptosis after Fas stimulation. GSH monoethyl ester is also protective, suggesting possible clinical applications.


Assuntos
Apoptose/fisiologia , Proteína Ligante Fas/metabolismo , Dissulfeto de Glutationa/metabolismo , Glutationa/deficiência , Falência Hepática Aguda/dietoterapia , Falência Hepática Aguda/metabolismo , Fígado/metabolismo , Aminoácidos Sulfúricos/administração & dosagem , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Dieta , Suplementos Nutricionais , Regulação para Baixo , Proteína Ligante Fas/agonistas , Glutationa/antagonistas & inibidores , Glutationa/farmacologia , Dissulfeto de Glutationa/farmacologia , Cetonas/administração & dosagem , Fígado/ultraestrutura , Falência Hepática Aguda/induzido quimicamente , Masculino , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Mitocôndrias Hepáticas/efeitos dos fármacos , NADPH Oxidases/metabolismo , Oxirredução , Fosforilação , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
9.
J Pharmacol Exp Ther ; 318(1): 444-54, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16617166

RESUMO

Like other nonsteroidal anti-inflammatory drugs, nimesulide (4-nitro-2-phenoxymethane-sulfoanilide) triggers hepatitis in a few recipients. Although nimesulide has been shown to uncouple mitochondrial respiration and cause hepatocyte necrosis in the absence of albumin, mechanisms for cell death are incompletely understood, and comparisons with human concentrations are difficult because 99% of nimesulide is albumin-bound. We studied the effects of nimesulide, with or without a physiological concentration of albumin, in isolated rat liver mitochondria or microsomes and in human hepatoma cells. Nimesulide did not undergo monoelectronic nitro reduction in microsomes. In mitochondria incubated without albumin, nimesulide (50 microM) decreased the mitochondrial membrane potential (DeltaPsim), increased basal respiration, and potentiated the mitochondrial permeability transition (MPT) triggered by calcium preloading. In HUH-7 cells incubated for 24 h without albumin, nimesulide (1 mM) decreased the DeltaPsim and cell NADPH and increased the glutathione disulfide/reduced glutathione ratio and cell peroxides; nimesulide triggered MPT, ATP depletion, high cell calcium, and caused mostly necrosis, with rare apoptotic cells. Coincubation with either cyclosporin A (an MPT inhibitor) or the combination of fructose-1,6-diphosphate (a glycolysis substrate) and oligomycin (an ATPase inhibitor) prevented the decrease in DeltaPsim, ATP depletion, and cell death. A physiological concentration of albumin abolished the effects of nimesulide on isolated mitochondria or HUH-7 cells. In conclusion, the weak acid, nimesulide, uncouples mitochondria and triggers MPT and ATP depletion in isolated mitochondria or hepatoma cells incubated without albumin. However, in the presence of albumin, only a fraction of the drug enters cells or organelles, and uncoupling and toxicity are not observed.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Carcinoma Hepatocelular/metabolismo , Citoproteção/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Soroalbumina Bovina/farmacologia , Sulfonamidas/farmacologia , Animais , Anti-Inflamatórios não Esteroides/química , Carcinoma Hepatocelular/patologia , Bovinos , Linhagem Celular Tumoral , Citoproteção/fisiologia , Humanos , Masculino , Camundongos , Mitocôndrias Hepáticas/metabolismo , Permeabilidade/efeitos dos fármacos , Ratos , Sulfonamidas/química , Desacopladores/química , Desacopladores/farmacologia
10.
J Hepatol ; 39(2): 171-8, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12873812

RESUMO

BACKGROUND/AIMS: Several cytochromes P450 (CYPs) are expressed in differentiated hepatocytes, but downregulated in growth-stimulated cells. We determined the signals involved in CYP downregulation by epidermal growth factor (EGF). METHODS: Rat hepatocytes were cultured with or without diverse substances for 72 h and EGF for the last 48 h. RESULTS: EGF increased c-myc mRNA and protein, and decreased CYP mRNAs and proteins; both effects were prevented by two agents blocking c-myc transcription (retinoic acid and DMSO) and two antisense c-myc oligomers. Despite unchanged CCAAT-enhancer binding protein alpha (C/EBPalpha) and increased C/EBPbeta levels, nuclear proteins of EGF-treated cells did not bind to a C/EBP DNA probe in a gel mobility shift assay. This binding was restored when cells were co-treated with both EGF and c-myc antisense oligomers (preventing c-Myc induction). The N-terminal c-Myc domain added to control nuclear extracts prevented C/EBP DNA binding. A monoclonal anti-c-Myc antibody co-immunoprecipitated c-Myc, C/EBPalpha and C/EBPbeta from nuclear extracts. In cells not treated with EGF, an antisense C/EBPalpha oligomer decreased CYP expression. CONCLUSIONS: EGF overexpresses c-Myc, decreases C/EBP binding to DNA and downregulates CYPs. We suggest that c-Myc may form inactive complexes with C/EBPs, thus decreasing C/EBP-mediated CYP transactivation.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Hepatócitos/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Antineoplásicos/farmacologia , Diferenciação Celular/fisiologia , Células Cultivadas , Dimetil Sulfóxido/farmacologia , Regulação para Baixo/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Sequestradores de Radicais Livres/farmacologia , Expressão Gênica/efeitos dos fármacos , Hepatócitos/citologia , Masculino , Oligonucleotídeos Antissenso , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Tretinoína/farmacologia
11.
Hepatology ; 39(3): 655-66, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14999684

RESUMO

The hepatotoxicity of several drugs is increased by mild viral infections. During such infections, death receptor ligands are expressed at low levels, and most parenchymal cells survive. We tested the hypothesis that subliminal death receptor stimulation may aggravate the hepatotoxicity of drugs, which are transformed by cytochrome P-450 cytochrome P-450 into glutathione-depleting reactive metabolites. Twenty-four-hour-fasted mice were pretreated with a subtoxic dose of the agonistic Jo2 anti-Fas antibody (1 microg per mouse) 3 hours before acetaminophen (500 mg/kg) or 1 hour before bromobenzene (400 mg/kg) administration. Administration of Jo2 alone increased hepatic inducible nitric oxide synthase nitric oxide synthase but did not modify serum alanine aminotransferase (ALT), hepatic adenosine triphosphate (ATP), glutathione (GSH), cytochrome P-450, cytosolic cytochrome c, caspase-3 activity or hepatic morphology. However, pretreating mice with Jo2 further decreased both hepatic GSH and ATP by 40% 4 hours after acetaminophen administration, and further increased serum ALT and the area of centrilobular necrosis at 24 hours. In mice pretreated with the Jo2 antibody before bromobenzene administration, hepatic GSH 4 hours after bromobenzene administration was 51% lower than in mice treated with bromobenzene alone, and serum ALT activity at 24 hours was 47-fold higher. In conclusion, administration of a subtoxic dose of an agonistic anti-Fas antibody before acetaminophen or bromobenzene increases metabolite-mediated GSH depletion and hepatotoxicity. Subliminal death receptor stimulation may be one mechanism whereby mild viral infections can increase drug-induced toxicity.


Assuntos
Acetaminofen/intoxicação , Analgésicos não Narcóticos/intoxicação , Bromobenzenos/intoxicação , Fígado/efeitos dos fármacos , Receptor fas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Caspase 3 , Caspases/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Glutamato-Cisteína Ligase/metabolismo , Glutationa/antagonistas & inibidores , Fígado/metabolismo , Hepatopatias/etiologia , Hepatopatias/mortalidade , Masculino , Camundongos , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II
12.
Hepatology ; 38(3): 715-25, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12939598

RESUMO

After several weeks of treatment, levels of alanine aminotransferase (ALT) increase in 50% of patients treated with tacrine for Alzheimer's disease. We looked for progressive effects on DNA to explain delayed toxicity. We first studied the in vitro effects of tacrine on DNA replication and topoisomerase-mediated DNA relaxation. We then treated mice with doses of tacrine reproducing the human daily dose on a body area basis and studied the effects of tacrine administration for up to 28 days on hepatic DNA, mitochondrial function, and cell death. In vitro, tacrine impaired DNA polymerase gamma-mediated DNA replication and also poisoned topoisomerases I and II to increase the relaxation of a supercoiled plasmid. In vivo, administration of tacrine markedly decreased incorporation of [(3)H]thymidine into mitochondrial DNA (mtDNA), progressively and severely depleted mtDNA, and partly unwound supercoiled mtDNA into circular mtDNA. Incorporation of [(3)H]thymidine into nuclear DNA (nDNA) was barely decreased, and nDNA levels were unchanged. After 12 to 28 days of treatment, administration of tacrine increased p53, Bax, mitochondrial permeability transition, cytosolic cytochrome c, and caspase-3 activity and triggered hepatocyte apoptosis and/or necrosis. In conclusion, the intercalating drug tacrine poisons topoisomerases and impairs DNA synthesis. Tacrine has been shown to accumulate within mitochondria, and it particularly targets mtDNA. After several weeks of treatment, the combination of severe mtDNA depletion and a genotoxic stress enhancing p53, Bax, and permeability transition trigger hepatocyte necrosis and/or apoptosis.


Assuntos
DNA Mitocondrial/metabolismo , DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Fígado/efeitos dos fármacos , Fígado/fisiologia , Tacrina/farmacologia , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Animais , Apoptose , DNA/biossíntese , Fragmentação do DNA , DNA Polimerase gama , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/fisiologia , DNA Circular/biossíntese , DNA Mitocondrial/efeitos dos fármacos , DNA Super-Helicoidal/efeitos dos fármacos , DNA Super-Helicoidal/fisiologia , DNA Polimerase Dirigida por DNA/fisiologia , Inibidores Enzimáticos/intoxicação , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Necrose , Estresse Oxidativo , Consumo de Oxigênio/efeitos dos fármacos , Permeabilidade , Tacrina/administração & dosagem , Tacrina/intoxicação , Timidina/antagonistas & inibidores , Timidina/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA