Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 12(4): 651-6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22310326

RESUMO

Arsenite is critical pharmacologically as a treatment for advanced stage blood cancer. However, environmental exposure to arsenic results in multiple diseases. Previous studies have shown that arsenic decreases expression of CYP3A, a critical drug metabolizing enzyme in human and rat liver. In addition, acute and chronic arsenic exposure in liver stimulates an inflammatory response. Our work has shown that arsenite decreases nuclear levels of RXRα the nuclear receptor that, as a heterodimer partner with PXR, transactivates the CYP3A gene. These results suggest that arsenite decreases transcription of CYP3A by decreasing RXRα. The present report shows that exposure to 5 µM arsenite decreased the activity of a rat CYP3A promoter luciferase reporter in HepG2 cells. The activity of a RARE-luciferase reporter, that is transactivated by the retinoic acid receptor (RAR)/RXRα, was also decreased. Previous studies have shown that arsenic in the concentration range of 2-5 µM affects CYP3A mRNA. When rifampicin-treated primary human hepatocyte cultures were exposed to arsenite concentrations as low as 50 nM, CYP3A mRNA was decreased. Treatment of primary human hepatocytes with the proteasome inhibitor MG132 increased RXRα suggesting the involvement of the proteasome pathway in regulation of RXRα. Finally, arsenic induces a pro-inflammatory response in liver. Surprisingly, we show that in hepatocytes arsenite decreases expression of two inflammatory mediators, TNF and VEGF, an effect that is not predicted from suppression of RXRα activity.


Assuntos
Arsenitos/toxicidade , Citocromo P-450 CYP3A/genética , Poluentes Ambientais/toxicidade , Hepatócitos/efeitos dos fármacos , Receptor X Retinoide alfa/metabolismo , Adulto , Células Cultivadas , Feminino , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Receptor de Pregnano X , RNA Mensageiro/metabolismo , Receptores de Esteroides/metabolismo , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator A de Crescimento do Endotélio Vascular/genética
2.
Hepatology ; 46(6): 1927-834, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17854053

RESUMO

UNLABELLED: Porphyria cutanea tarda is a liver disease characterized by elevated hepatic iron and excessive production of uroporphyrin (URO). Phlebotomy is an effective treatment that probably acts by reducing hepatic iron. Here we used Hfe(-/-) mice to compare the effects on hepatic URO accumulation of two different methods of hepatic iron depletion: iron chelation using deferiprone (L1) versus iron-deficient diets. Hfe(-/-) mice in a 129S6/SvEvTac background were fed 5-aminolevulinic acid (ALA), which results in hepatic URO accumulation, and increasing doses of L1 in the drinking water. Hepatic URO accumulation was completely prevented at low L1 doses, which partially depleted hepatic nonheme iron. By histological assessment, the decrease in hepatic URO accumulation was associated with greater depletion of iron from hepatocytes than from Kupffer cells. The L1 treatment had no effect on levels of hepatic cytochrome P4501A2 (CYP1A2). L1 also effectively decreased hepatic URO accumulation in C57BL/6 Hfe(-/-) mice treated with ALA and a CYP1A2 inducer. ALA-treated mice maintained on defined iron-deficient diets, rather than chow diets, did not develop uroporphyria, even when the animals were iron-supplemented either directly in the diet or by iron dextran injection. CONCLUSION: The results suggest that dietary factors other than iron are involved in the development of uroporphyria and that a modest depletion of hepatocyte iron by L1 is sufficient to prevent URO accumulation.


Assuntos
Quelantes de Ferro/uso terapêutico , Deficiências de Ferro , Porfiria Cutânea Tardia/dietoterapia , Porfiria Cutânea Tardia/tratamento farmacológico , Piridonas/uso terapêutico , Animais , Deferiprona , Modelos Animais de Doenças , Fígado/química , Masculino , Camundongos , Uroporfirinas/análise
3.
Hepatology ; 45(1): 187-94, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17187429

RESUMO

UNLABELLED: Excess hepatic iron is known to enhance both porphyria cutanea tarda (PCT) and experimental uroporphyria. Since previous studies have suggested a role for ascorbate (AA) in suppressing uroporphyria in AA-requiring rats (in the absence of excess iron), the present study investigated whether AA could suppress uroporphyria produced by excess hepatic iron. Hepatic URO accumulation was produced in AA-requiring Gulo(-/-) mice by treatment with 3,3',4,4',5-pentachlorbiphenyl, an inducer of CYP1A2, and 5-aminolevulinic acid. Mice were administered either sufficient AA (1000 ppm) in the drinking water to maintain near normal hepatic AA levels or a lower intake (75 ppm) that resulted in 70 % lower hepatic AA levels. The higher AA intake suppressed hepatic URO accumulation in the absence of administered iron, but not when iron dextran (300-500 mg Fe/kg) was administered. This effect of iron was not due to hepatic AA depletion since hepatic AA content was not decreased. The effect of iron to prevent AA suppression of hepatic URO accumulation was not observed until a high hepatic iron threshold was exceeded. At both low and high AA intakes, hepatic malondialdehyde (MDA), an indicator of oxidative stress, was increased three-fold by high doses of iron dextran. MDA was considerably increased even at low iron dextran doses, but without any increase in URO accumulation. The level of hepatic CYP1A2 was unaffected by either AA intake. CONCLUSION: In this mouse model of PCT, AA suppresses hepatic URO accumulation at low, but not high hepatic iron levels. These results may have implications for the management of PCT.


Assuntos
Deficiência de Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Ferro/farmacologia , Porfiria Cutânea Tardia/metabolismo , Uroporfirinas/metabolismo , Ácido Aminolevulínico , Animais , Ácido Ascórbico/genética , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ferro/metabolismo , Complexo Ferro-Dextran/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Bifenilos Policlorados , Porfiria Cutânea Tardia/induzido quimicamente , Porfiria Cutânea Tardia/tratamento farmacológico
4.
Toxicol Appl Pharmacol ; 217(3): 245-51, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17083955

RESUMO

Previous work in our laboratory has shown that acute exposure of primary rat hepatocyte cultures to non-toxic concentrations of arsenite causes major decreases in the DEX-mediated induction of CYP3A23 protein, with minor decreases in CYP3A23 mRNA. To elucidate the mechanism for these effects of arsenite, the effects of arsenite and proteasome inhibition, separately and in combination, on induction of CYP3A23 protein were compared. The proteasome inhibitor, MG132, inhibited proteasome activity, but also decreased CYP3A23 mRNA and protein. Lactacystin, another proteasome inhibitor, decreased CYP3A23 protein without affecting CYP3A23 mRNA at a concentration that effectively inhibited proteasome activity. This result, suggesting that the action of lactacystin is similar to arsenite and was post-transcriptional, was confirmed by the finding that lactacystin decreased association of DEX-induced CYP3A23 mRNA with polyribosomes. Both MG132 and lactacystin inhibited total protein synthesis, but did not affect MTT reduction. Arsenite had no effect on ubiquitination of proteins, nor did arsenite significantly affect proteasomal activity. These results suggest that arsenite and lactacystin act by similar mechanisms to inhibit translation of CYP3A23.


Assuntos
Arsenitos/toxicidade , Hidrocarboneto de Aril Hidroxilases/biossíntese , Hepatócitos/efeitos dos fármacos , Inibidores de Proteassoma , Acetilcisteína/análogos & derivados , Acetilcisteína/toxicidade , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Células Cultivadas , Inibidores de Cisteína Proteinase/toxicidade , Citocromo P-450 CYP3A , Dexametasona/farmacologia , Interações Medicamentosas , Hepatócitos/enzimologia , Leupeptinas/toxicidade , Masculino , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344
5.
Am J Physiol Gastrointest Liver Physiol ; 290(6): G1269-79, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16439473

RESUMO

The objective of this study was to determine whether Toll-like receptor 4 (TLR4) has a role in alcohol-mediated acetaminophen (APAP) hepatotoxicity. TLR4 is involved in the inflammatory response to endotoxin. Others have found that ethanol-mediated liver disease is decreased in C3H/HeJ mice, which have a mutated TLR4 resulting in a decreased response to endotoxin compared with endotoxin-responsive mice. In the present study, short-term (1 wk) pretreatment with ethanol plus isopentanol, the predominant alcohols in alcoholic beverages, caused no histologically observed liver damage in either C3H/HeJ mice or endotoxin-responsive C3H/HeN mice, despite an increase in nitrotyrosine levels in the livers of C3H/HeN mice. In C3H/HeN mice pretreated with the alcohols, subsequent exposure to APAP caused a transient decrease in liver nitrotyrosine formation, possibly due to competitive interaction of peroxynitrite with APAP producing 3-nitroacetaminophen. Treatment with APAP alone resulted in steatosis in addition to congestion and necrosis in both C3H/HeN and C3H/HeJ mice, but the effects were more severe in endotoxin-responsive C3H/HeN mice. In alcohol-pretreated endotoxin-responsive C3H/HeN mice, subsequent exposure to APAP resulted in further increases in liver damage, including severe steatosis, associated with elevated plasma levels of TNF-alpha. In contrast, alcohol pretreatment of C3H/HeJ mice caused little to no increase in APAP hepatotoxicity and no increase in plasma TNF-alpha. Portal blood endotoxin levels were very low and were not detectably elevated by any of the treatments. In conclusion, this study implicates a role of TLR4 in APAP-mediated hepatotoxicity.


Assuntos
Acetaminofen/efeitos adversos , Etanol/efeitos adversos , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Receptor 4 Toll-Like/metabolismo , Analgésicos não Narcóticos/efeitos adversos , Animais , Sinergismo Farmacológico , Fígado Gorduroso/patologia , Feminino , Fígado/patologia , Camundongos
6.
Biochem Biophys Res Commun ; 333(4): 1211-7, 2005 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15979568

RESUMO

In primary cultures of rat hepatocytes, exposure to arsenite causes a major decrease in dexamethasone (DEX)-mediated induction of CYP3A23 hemoprotein, with a minor decrease in CYP3A23 mRNA. Here we show that addition of heme did not prevent the arsenite-mediated decreases in CYP3A23 protein, and arsenite did not decrease intracellular glutathione levels, indicating that heme and glutathione were not limiting for formation of holoCYP3A23. We also investigated whether arsenite decreases CYP3A23 protein by increasing CYP3A23 degradation by the calpain pathway. The calpain inhibitor, calpeptin, caused greater than a 90% inhibition of calpain-mediated proteolysis, but had no effect on DEX-mediated induction of CYP3A23 protein following 24h treatments. However, calpeptin enhanced the effect of arsenite to decrease induction of CYP3A23 protein. In addition, in short-term studies, calpeptin appeared to be a suicidal inhibitor of CYP3A-catalyzed enzyme activity. Our findings suggest that CYP3A23 protein is not degraded by calpain-mediated proteolysis, even in the presence of arsenite.


Assuntos
Arsenitos/administração & dosagem , Hidrocarboneto de Aril Hidroxilases/metabolismo , Calpaína/metabolismo , Glutationa/metabolismo , Heme/metabolismo , Hepatócitos/metabolismo , Animais , Células Cultivadas , Citocromo P-450 CYP3A , Dipeptídeos/farmacologia , Relação Dose-Resposta a Droga , Hepatócitos/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos F344 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
7.
Arch Biochem Biophys ; 439(1): 1-11, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15946643

RESUMO

Primary cultures of chick embryo hepatocytes have been used to study the mechanisms by which various drugs and other chemicals cause accumulation of porphyrin intermediates of the heme pathway. When these cultures are incubated with the heme precursor, 5-aminolevulinic acid (ALA), there is a major accumulation of protoporphyrin. However, in the presence of ALA, addition of insulin caused a striking increase in accumulation of uroporphyrin I and coproporphyrin III, whereas addition of glucagon mainly caused an increase in uroporphyrin I. Treatment with both insulin and glucagon resulted in additive increases in uroporphyrin, but not coproporphyrin. Antioxidants abolished the uroporphyrin I accumulation and increased coproporphyrin III. Insulin caused an increase in uptake of ALA and an increase in porphobilinogen accumulation, suggesting that the accumulation of uroporphyrin I is due to increased flux through the heme pathway. Apparently, this increased flux could particularly affect the utilization of the intermediate hydroxymethylbilane, which would result in accumulation of uroporphyrin I.


Assuntos
Ácido Aminolevulínico/metabolismo , Coproporfirinas/biossíntese , Fármacos Gastrointestinais/farmacologia , Glucagon/farmacologia , Hepatócitos/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Uroporfirinas/biossíntese , Ácido Aminolevulínico/farmacologia , Animais , Células Cultivadas , Embrião de Galinha , Hepatócitos/efeitos dos fármacos , Fármacos Fotossensibilizantes/farmacologia
8.
Toxicol Appl Pharmacol ; 209(2): 174-82, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15907335

RESUMO

Arsenic is a naturally occurring, worldwide contaminant implicated in numerous pathological conditions in humans, including cancer and several forms of liver disease. One of the contributing factors to these disorders may be the alteration of cytochrome P450 (CYP) levels by arsenic. In rat and human hepatocyte cultures, arsenic, in the form of arsenite, decreases the induction of several CYPs. The present study investigated whether arsenite utilizes transcriptional or post-transcriptional mechanisms to decrease CYP3A23 in primary cultures of rat hepatocytes. In these cultures, a 6-h treatment with 5 microM arsenite abolished dexamethasone (DEX)-mediated induction of CYP3A23 protein and activity, but did not inhibit general protein synthesis. However, arsenite treatment only reduced DEX-induced levels of CYP3A23 mRNA by 30%. The effects of arsenite on CYP3A23 transcription were examined using a luciferase reporter construct containing 1.4 kb of the CYP3A23 promoter. Arsenite caused a 30% decrease in DEX-induced luciferase expression of this reporter. Since arsenite abolished induction of CYP3A23 protein, but caused only a small decrease in CYP3A23 mRNA, the effects of arsenite on translation of CYP3A23 mRNA were investigated. Polysomal distribution analysis showed that arsenite decreased translation by decreasing the DEX-mediated increase in CYP3A23 mRNA association with polyribosomes. Arsenite did not decrease intracellular glutathione or increase lipid peroxidation, suggesting that the effect of arsenite on CYP3A23 does not involve oxidative stress. Overall, the results suggest that low-level arsenite decreases both transcription and translation of CYP3A23 in primary rat hepatocyte cultures.


Assuntos
Arsenitos/toxicidade , Hidrocarboneto de Aril Hidroxilases/biossíntese , Fígado/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Animais , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/genética , Citocromo P-450 CYP3A , Dexametasona/farmacologia , Indução Enzimática/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Immunoblotting , Fígado/enzimologia , Fígado/metabolismo , Masculino , Polirribossomos/enzimologia , Polirribossomos/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Drug Metab Dispos ; 33(7): 993-1003, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15833926

RESUMO

Arsenic is a naturally occurring, worldwide contaminant implicated in numerous pathological conditions in humans, including cancer and several forms of liver disease. One of the contributing factors to these disorders may be the alteration of cytochrome P450 (P450) levels by arsenic. P450s are involved in the oxidative metabolism and elimination of numerous toxic chemicals. CYP3A4, a major P450 in humans, is involved in the metabolism of half of all currently used drugs. Acute exposure to arsenite decreases the induction of CYP1A1/2 proteins and activities in cultured human hepatocytes, as well as CYP3A23 in cultured rat hepatocytes. Here, in primary cultures of human hepatocytes, we assessed the effects of acute arsenite exposure on CYP3A4 and several transcription factors involved in CYP3A4 expression. The concentrations of arsenite used in these studies were nontoxic to the hepatocytes and failed to elicit an oxidative response. Treatment with arsenite in the presence of CYP3A4 inducers, rifampicin (Rif) or phenobarbital, caused major decreases in CYP3A4 mRNA, protein, and activity. In addition, the levels of CYP3A4 in untreated cells were decreased following arsenite treatment. Transcription of the CYP3A4 gene is primarily regulated by heterodimers of the retinoid X receptor alpha (RXRalpha) and the pregnane X receptor (PXR). We found that arsenite failed to affect expression of PXR or the transcription factor Sp1, yet caused a significant decrease in PXR responsiveness to Rif. Arsenite caused a large decrease in nuclear RXRalpha protein and, to a lesser extent, RXRalpha mRNA. These results suggest that arsenite inhibits both untreated and induced CYP3A4 transcription in primary human hepatocytes by decreasing the activity of PXR, as well as expression of the nuclear receptor RXRalpha.


Assuntos
Arsenitos/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Hepatócitos/efeitos dos fármacos , Receptor X Retinoide alfa/antagonistas & inibidores , Adolescente , Adulto , Idoso , Sequência de Bases , Células Cultivadas , Criança , Pré-Escolar , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Primers do DNA , Feminino , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética
10.
Hepatology ; 40(4): 942-50, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15382179

RESUMO

Two major risk factors for porphyria cutanea tarda (PCT) are alcohol consumption and homozygosity for the C282Y mutation in the hereditary hemochromatosis gene (HFE). We recently described an animal model for alcohol-induced uroporphyria, using Hfe(-/-) mice. In the present study we show that this effect is dependent on genetic background and ethanol dose. In the 129S6/SvEvTac (129) strain, treatment with 15% ethanol in the drinking water for 6.5 months produced an accumulation of hepatic uroporphyrin (URO) 4-fold higher than that observed with 10% ethanol, a 90% decrease in uroporphyrinogen decarboxylase activity (UROD), and further increased the activities of hepatic 5-aminolevulinate synthase (ALAS) and CYP1A2. Hepatic nonheme iron (NHFe) and hepatocyte iron staining were not further increased by 15% compared to 10% ethanol. Treatment of C57BL/6 Hfe(-/-) mice with 15% ethanol for 6.5 months did not increase hepatic URO. Although NHFe was increased by ethanol, the resulting level was only half that of ethanol-treated 129 Hfe(-/-) mice. ALAS induction was similar in both Hfe(-/-) strains. In wild-type 129 mice treated with ethanol for 6 to 7 months, administration of iron dextran increased hepatic URO accumulation and decreased UROD activity. In conclusion, this study demonstrates a strong effect of genetic background on ethanol-induced uroporphyria, which is probably due to a greater effect of ethanol on iron metabolism in the susceptible strain.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Antígenos de Histocompatibilidade Classe I/genética , Proteínas de Membrana/genética , Porfiria Cutânea Tardia/genética , 5-Aminolevulinato Sintetase/metabolismo , Consumo de Bebidas Alcoólicas/metabolismo , Animais , Citocromo P-450 CYP1A2/metabolismo , Proteína da Hemocromatose , Ferro/metabolismo , Complexo Ferro-Dextran/farmacologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Porfiria Cutânea Tardia/etiologia , Porfiria Cutânea Tardia/metabolismo , Uroporfirinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...