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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 266(3): 419-29, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23231920

RESUMO

In the current study C57BL/6J mice were injected intraperitoneally with Hg(2+) in the absence and presence of TCDD. After 6 and 24h the liver was harvested and the expression of Cyps was determined. In vitro, isolated hepatocytes were incubated with TCDD in the presence and absence of Hg(2+). At the in vivo level, Hg(2+) significantly decreased the TCDD-mediated induction of Cyps at 6h while potentiating their levels at 24h. In vitro, Hg(2+) significantly inhibited the TCDD-mediated induction of Cyp1a1 in a concentration- and time-dependent manner. Interestingly, Hg(2+) increased the serum hemoglobin (Hb) levels in mice treated for 24h. Upon treatment of isolated hepatocytes with Hb alone, there was an increase in the AhR-dependent luciferase activity with a subsequent increase in Cyp1a1 protein and catalytic activity levels. Importantly, when hepatocytes were treated for 2h with Hg(2+) in the presence of TCDD, then the medium was replaced with new medium containing Hb, there was potentiation of the TCDD-mediated effect. In addition, Hg(2+) increased heme oxygenase-1 (HO-1) mRNA, which coincided with a decrease in the Cyp1a1 activity level. When the competitive HO-1 inhibitor, tin mesoporphyrin was applied to the hepatocytes there was a partial restoration of Hg(2+)-mediated inhibition of Cyp1a1 activity. In conclusion, we demonstrate for the first time that there is a differential modulation of the TCDD-mediated induction of Cyp1a1 by Hg(2+) in C57BL/6J mice livers and isolated hepatocytes. Moreover, this study implicates Hb as an in vivo specific modulator of Cyp1 family.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Heme Oxigenase-1/metabolismo , Fígado/efeitos dos fármacos , Mercúrio/toxicidade , Dibenzodioxinas Policloradas/toxicidade , Animais , Western Blotting , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/genética , Hemoglobinas/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Masculino , Metaloporfirinas/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , RNA/química , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real
2.
Arch Toxicol ; 87(8): 1531-43, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23423713

RESUMO

The objective of the current study was to investigate the effect of vanadium (V(5+)) on Cyp1 expression and activity in C57BL/6 mice liver and isolated hepatocytes. For this purpose, C57BL6 mice were injected intraperitoneally with V(5+) (5 mg/kg) in the absence and presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (15 µg/kg) for 6 and 24 h. Furthermore, isolated hepatocytes from C57BL6 mice were treated with V(5+) (5, 10, and 20 µM) in the absence and presence of TCDD (1 nM) for 3, 6, 12, and 24 h. In vivo, V(5+) alone did not significantly alter Cyp1a1, Cyp1a2, or Cyp1b1 mRNA, protein, or catalytic activity levels. Upon co-exposure to V(5+) and TCDD, V(5+) significantly potentiated the TCDD-mediated induction of the Cyp1a1, Cyp1a2, and Cyp1b1 mRNA, protein, and catalytic activity levels at 24 h. In vitro, V(5+) decreased the TCDD-mediated induction of Cyp1a1 mRNA, protein, and catalytic activity levels. Furthermore, V(5+) significantly inhibited the TCDD-induced AhR-dependent luciferase activity. V(5+) also increased serum hemoglobin (Hb) levels in animals treated for 24 h. Upon treatment of isolated hepatocytes with Hb alone or in the presence of TCDD, there was an increase in the AhR-dependent luciferase activity. When isolated hepatocytes were treated for 2 h with V(5+) in the presence of TCDD, followed by replacement of the medium with new medium containing Hb, there was further potentiation to the TCDD-mediated effect. The present study demonstrates that there is a differential modulation of Cyp1a1 by V(5+) in C57BL/6 mice livers and isolated hepatocytes and demonstrates Hb as an in vivo specific modulator.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Dibenzodioxinas Policloradas/farmacologia , Vanádio/farmacologia , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1B1 , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica , Heme Oxigenase-1/genética , Hemoglobinas/metabolismo , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Hidrocarboneto Arílico/metabolismo
3.
J Appl Toxicol ; 33(11): 1230-40, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22611038

RESUMO

We recently reported that vanadium (V(5+) ) was able to decrease the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated induction of Cyp1a1 and Nqo1 at mRNA, protein and catalytic activity levels in mouse hepatoma Hepa 1c1c7 and human hepatoma HepG2 cells. However, little is known regarding the in vivo effects. Thus, the objective of this study was to investigate whether similar effects would occur at the in vivo level. Therefore, we examined the effect of exposure to V(5+) (5 mg kg(-1) ) with or without TCDD (15 µg kg(-1) ) on the AhR-regulated genes in kidney, lung and heart of C57BL/6 J mice. Our results demonstrated that V(5+) alone significantly decreased Cyp1b1 protein and catalytic activity levels in kidney at 24 h. Moreover, it significantly potentiated Nqo1 and Gsta1 gene expression in the heart, and only Gsta1 gene expression in the lung. Upon co-exposure, we found that V(5+) significantly inhibited the TCDD-mediated induction of Cyp1a1, Cyp1a2 and Cyp1b1 mRNA, protein and catalytic activity levels in the kidney at 24 h. On the other hand, V(5+) significantly potentiated the TCDD-mediated induction of Nqo1 and Gsta1 protein and activity levels in the kidney. Cyp1a1, Cyp1b1, Nqo1 mRNA, protein and catalytic activity levels in the lung were significantly potentiated at 6 h. Interestingly, all tested genes in the heart were significantly decreased at 6 h with the exception of Gsta1 mRNA. The present study demonstrates that V(5+) modulates TCDD-induced AhR-regulated genes. Furthermore, the effect on one of these enzymes could not be generalized to other enzymes even if it was in the same organ.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Rim/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/genética , Vanadatos/farmacologia , Animais , Western Blotting , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1B1/genética , Interações Medicamentosas , Glutationa Transferase/genética , Isoenzimas/genética , Rim/metabolismo , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microssomos/efeitos dos fármacos , Microssomos/metabolismo , Miocárdio/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Reação em Cadeia da Polimerase em Tempo Real
4.
Arch Toxicol ; 86(6): 897-910, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22535251

RESUMO

During the last couple of decades, efforts have been made to study the toxic effects of individual aryl hydrocarbon receptors (AhR) ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or heavy metals typified by arsenic As(III). However, little is known about the combined toxic effects of TCDD and As(III) in vivo. Previous reports from our laboratory and others have demonstrated that As(III), by itself or in the presence of AhR ligands, such as TCDD, is capable of differentially altering the expression of various phase I and phase II AhR-regulated genes in in vitro systems. Thus, the objective of the current study was to investigate whether or not similar effects would occur at the in vivo level. Therefore, we examined the effect of exposure to As(III) (12.5 mg/kg) in the absence and presence of TCDD (15 µg/kg) on the AhR-regulated genes using C57Bl/6 mice. Our results demonstrated that As(III) alone inhibited Cyp1a1 and Cyp1a2 in the kidney, while it induced their levels in the lung and did not affect their mRNA levels in the heart. As(III) also induced Nqo1 and Gsta1 in all tested tissues. Upon co-exposure to As(III) and TCDD, As(III) inhibited the TCDD-mediated induction of Cyp1a1 in the kidney and heart, Cyp1a2 in the kidney and heart, while it potentiated TCDD-mediated induction of Cyp1a1 in the lung, and Nqo1 and Gsta1 in the kidney and lung. In conclusion, the present study demonstrates for the first time that As(III) modulates constitutive and TCDD-induced AhR-regulated genes in a time-, tissue-, and AhR-regulated enzyme-specific manner.


Assuntos
Arsenitos/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/metabolismo , Pulmão/metabolismo , Miocárdio/metabolismo , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/genética , Animais , Misturas Complexas/toxicidade , Ativação Enzimática/efeitos dos fármacos , Rim/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dibenzodioxinas Policloradas/toxicidade
5.
Toxicol Lett ; 226(1): 53-62, 2014 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-24472606

RESUMO

Mercury exposure is associated with increased risk of cardiovascular disease and profound cardiotoxicity. However, the correlation between Hg(2+)-mediated toxicity and alteration in cardiac cytochrome P450s (Cyp) and their dependent arachidonic acid metabolites has never been investigated. Therefore, we investigated the effect of acute mercury toxicity on the expression of Cyp-epoxygenases and Cyp-ω-hydroxylases and their associated arachidonic acid metabolites in mice hearts. In addition, we examined the expression and activity of soluble epoxide hydrolase (sEH) as a key player in arachidonic acid metabolism pathway. Mercury toxicity was induced by a single intraperitoneal injection (IP) of 2.5 mg/kg of mercuric chloride (HgCl2). Our results showed that mercury treatment caused a significant induction of the cardiac hypertrophy markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP); in addition to Cyp1a1, Cyp1b1, Cyp2b9, Cyp2b10, Cyp2b19, Cyp2c29, Cyp2c38, Cyp4a10, Cyp4a12, Cyp4a14, Cyp4f13, Cyp4f15, Cyp4f16 and Cyp4f18 gene expression. Moreover, Hg(2+) significantly increased sEH protein expression and activity levels in hearts of mercury-treated mice, with a consequent decrease in 14,15-, and 11,12-epoxyeicosatrienoic acids (EETs) levels. Whereas the formation of 14,15-, 11,12-, 8,9-dihydroxyeicosatrienoic acids (DHETs) was significantly increased. In conclusion, acute Hg(2+) toxicity modulates the expression of several Cyp and sEH enzymes with a consequent decrease in the cardioprotective EETs which could represent a novel mechanism by which mercury causes progressive cardiotoxicity. Furthermore, inhibiting sEH might represent a novel therapeutic approach to prevent Hg(2+)-induced hypertrophy.


Assuntos
Ácido Araquidônico/metabolismo , Cardiomegalia/induzido quimicamente , Sistema Enzimático do Citocromo P-450/metabolismo , Epóxido Hidrolases/metabolismo , Coração/efeitos dos fármacos , Cloreto de Mercúrio/toxicidade , Miocárdio/enzimologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animais , Fator Natriurético Atrial/metabolismo , Biomarcadores/metabolismo , Cardiomegalia/enzimologia , Cardiomegalia/genética , Sistema Enzimático do Citocromo P-450/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Isoenzimas , Masculino , Cloreto de Mercúrio/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Peptídeo Natriurético Encefálico/metabolismo , RNA Mensageiro/metabolismo
6.
Toxicol Lett ; 219(3): 239-47, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23541843

RESUMO

Recently we demonstrated the ability of mercuric chloride (Hg(2+)) in human hepatoma HepG2 cells to significantly decrease the TCDD-mediated induction of Cytochrome P450 1A1 (CYP1A1) mRNA, protein, and catalytic activity levels. In this study we investigated the effect of methylmercury (MeHg) on CYP1A1 in HepG2 cells. For this purpose, cells were co-exposed to MeHg and TCDD and the expression of CYP1A1 mRNA, protein, and catalytic activity levels were determined. Our results showed that MeHg did not alter the TCDD-mediated induction of CYP1A1 mRNA, or protein levels; however it was able to significantly decrease CYP1A1 catalytic activity levels in a concentration-dependent manner. Importantly, this inhibition was specific to CYP1A1and was not radiated to other aryl hydrocarbon receptor (AhR)-regulated genes, as MeHg induced NAD(P)H:quinone oxidoreductase 1 mRNA and protein levels. Mechanistically, the inhibitory effect of MeHg on the induction of CYP1A1 coincided with an increase in heme oxygenase-1 (HO-1) mRNA levels. Furthermore, the inhibition of HO-1 activity, by tin mesoporphyrin, caused a complete restoration of MeHg-mediated inhibition of CYP1A1 activity, induced by TCDD. In addition, transfection of HepG2 cells with siRNA targeting the human HO-1 gene reversed the MeHg-mediated inhibition of TCDD-induced CYP1A1. In conclusion, this study demonstrated that MeHg inhibited the TCDD-mediated induction of CYP1A1 through a posttranslational mechanism and confirms the role of HO-1 in a MeHg-mediated effect.


Assuntos
Citocromo P-450 CYP1A1/genética , Heme Oxigenase-1/metabolismo , Compostos de Metilmercúrio/farmacologia , Western Blotting , Citocromo P-450 CYP1A1/antagonistas & inibidores , Citocromo P-450 CYP1A1/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Heme Oxigenase-1/fisiologia , Células Hep G2/efeitos dos fármacos , Células Hep G2/metabolismo , Humanos , Dibenzodioxinas Policloradas/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real
7.
Free Radic Biol Med ; 58: 52-63, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23369756

RESUMO

Heavy metals, typified by arsenite (As(III)), have been implicated in altering the carcinogenicity of aryl hydrocarbon receptor (AhR) ligands, typified by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), by modulating the induction of the Cyp1a1 enzyme, but the mechanism remains unresolved. In this study, the effects of As(III) on Cyp1a1 expression and activity were investigated in C57BL/6 mouse livers and isolated hepatocytes. For this purpose, C57BL/6 mice were injected intraperitoneally with As(III) (12.5 mg/kg) in the absence and presence of TCDD (15 µg/kg) for 6 and 24 h. Furthermore, isolated hepatocytes from C57BL/6 mice were treated with As(III) (1, 5, and 10 µM) in the absence and presence of TCDD (1 nM) for 3, 6, 12, and 24 h. At the in vivo level, As(III) decreased the TCDD-mediated induction of Cyp1a1 mRNA at 6h while potentiating its mRNA, protein, and catalytic activity levels at 24 h. At the in vitro level, As(III) decreased the TCDD-mediated induction of Cyp1a1 mRNA in a concentration- and time-dependent manner. Moreover, As(III) decreased the TCDD-mediated induction of Cyp1a1 protein and catalytic activity levels at 24 h. Interestingly, As(III) increased the serum hemoglobin (Hb) levels in animals treated for 24 h. Upon treatment of isolated hepatocytes with Hb alone, there was an increase in the nuclear accumulation of AhR and AhR-dependent luciferase activity. Furthermore, Hb potentiated the TCDD-induced AhR-dependent luciferase activity. Importantly, when isolated hepatocytes were treated for 5h with As(III) in the presence of TCDD and the medium was then replaced with new medium containing Hb, there was potentiation of the TCDD-mediated effect. Taken together, these results demonstrate for the first time that there is a differential modulation of the TCDD-mediated induction of Cyp1a1 by As(III) in C57BL/6 mouse livers and isolated hepatocytes. Thus, this study implicates Hb as an in vivo-specific modulator.


Assuntos
Arsenitos/farmacologia , Citocromo P-450 CYP1A1/metabolismo , Hemoglobinas/metabolismo , Dibenzodioxinas Policloradas/farmacologia , Animais , Células Cultivadas , Radicais Livres/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Proteínas de Membrana/metabolismo , Metais Pesados/farmacologia , Camundongos , RNA Mensageiro/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/metabolismo
8.
Food Chem Toxicol ; 50(7): 2325-34, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22579925

RESUMO

The individual toxic effects of aryl hydrocarbon receptors (AhR) ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or heavy metals typified by mercury (Hg(2+)) has been previously demonstrated. However, little is known about the combined toxic effects of TCDD and Hg(2+)in vivo. Therefore, we examined the effect of exposure to Hg(2+) (2.5mg/kg) in the absence and presence of TCDD (15 µg/kg) on the AhR-regulated genes using C57Bl/6 mice. Hg(2+) alone did not affect kidney, lung, or heart Cyp1a1/1a2/1b1 mRNA levels. On the contrary, Hg(2+) alone significantly induced kidney Cyp1a1/1a2/1b1 and lung Cyp1b1 protein and catalytic activities. Hg(2+) also induced Nqo1, Gsta1, and HO-1 at the mRNA, protein, and activity levels in the kidney and heart but not in the lung. Upon co-exposure to Hg(2+) and TCDD, Hg(2+) significantly potentiated the TCDD-mediated induction of kidney and lung Cyp1a1/1a2/1b1 mRNA levels, while it decreased their kidney protein and catalytic activity and it increased their lung protein. In addition, Hg(2+) potentiated the TCDD-mediated induction of Nqo1, Gsta1, and HO-1 at mRNA, protein and activity levels in all tissues. The present study demonstrates that Hg(2+) modulates the constitutive and TCDD-induced AhR-regulated genes in a time-, tissue- and, AhR-regulated enzyme genes manner.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Mercúrio/toxicidade , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Animais , Sequência de Bases , Biocatálise , Western Blotting , Sistema Enzimático do Citocromo P-450/biossíntese , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Primers do DNA , Sinergismo Farmacológico , Indução Enzimática , Coração/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/enzimologia , Rim/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/enzimologia , Miocárdio/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Hidrocarboneto Arílico/fisiologia
9.
Free Radic Biol Med ; 51(9): 1675-85, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21843634

RESUMO

NAD(P)H:quinone oxidoreductase (NQO1)-mediated detoxification of quinones plays a critical role in cancer prevention. Heavy metals such as mercury (Hg(2+)) alter the carcinogenicity of aryl hydrocarbon receptor ligands, mainly by modifying various xenobiotic-metabolizing enzymes such as NQO1. Therefore, we examined the effect of Hg(2+) on the expression of NQO1 in human hepatoma HepG2 cells. For this purpose HepG2 cells were incubated with various concentrations of Hg(2+) (2.5, 5, and 10µM) in the presence and absence of two NQO1 inducers, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and isothiocyanate sulforaphane (SUL), as bifunctional and monofunctional inducers, respectively. Analysis of the time-dependent effect of Hg(2+) revealed that Hg(2+) increased the expression of NQO1 mRNA in a time-dependent manner. In addition, Hg(2+) increased NQO1 at the mRNA, protein, and activity levels in the presence and absence of both NQO1 inducers, TCDD and SUL, which coincided with increased nuclear accumulation of Nrf2 protein. Investigating the effect of Hg(2+) at the transcriptional level revealed that Hg(2+) significantly induced the antioxidant-responsive element-dependent luciferase reporter gene expression in the absence and the presence of both NQO1 inducers. NQO1 mRNA and protein decay experiments revealed a lack of posttranscriptional and posttranslational mechanisms. Transfecting HepG2 cells with siRNA for Nrf2 significantly decreased the Hg(2+)-mediated induction of NQO1 mRNA and catalytic activity by approximately 90%. In conclusion, we demonstrated that Hg(2+) regulates the expression of the NQO1 gene through a transcriptional mechanism in human hepatoma HepG2 cells. In addition, Nrf2 is involved in the modulation of NQO1 by Hg(2+).


Assuntos
Mercúrio/farmacologia , NAD(P)H Desidrogenase (Quinona)/genética , Transcrição Gênica/efeitos dos fármacos , Biocatálise , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Isotiocianatos/farmacologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , Dibenzodioxinas Policloradas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo
10.
Toxicol Lett ; 199(3): 225-33, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20837117

RESUMO

Aryl hydrocarbon receptor (AhR) ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and metals, such as mercury (Hg(2+)), are environmental co-contaminants and their molecular interaction may disrupt the coordinated regulation of the carcinogen-activating enzyme cytochrome P450 1A1 (CYP1A1). Therefore, we examined the effect of co-exposure to Hg(2+) and TCDD on the expression of the CYP1A1 in HepG2 cells. Our results showed that Hg(2+) significantly inhibited the TCDD-mediated induction of CYP1A1 at the mRNA, protein, and catalytic activity levels. At the transcriptional level, co-exposure to Hg(2+) and TCDD significantly decreased the TCDD-mediated induction of AhR-dependent luciferase reporter gene expression. Moreover, Hg(2+) did not affect CYP1A1 mRNA stability, while decreasing its protein half-life, suggesting the involvement of a posttranslational mechanism. Importantly, Hg(2+) increased the expression of heme oxygenase-1 (HO-1), a rate limiting enzyme in heme degradation, which coincided with further decrease in the CYP1A1 catalytic activity levels. Upon using a competitive HO-1 inhibitor, tin mesoporphyrin, heme precursor, hemin, or transfecting the HepG2 cells with siRNA for HO-1 there was a partial restoration of the inhibition of TCDD-mediated induction of CYP1A1 catalytic activity. In conclusion, we demonstrate that Hg(2+) down-regulates the expression of CYP1A1 at the transcriptional and posttranslational levels in HepG2 cells. In addition, HO-1 is involved in the modulation of CYP1A1 at the posttranslational level.


Assuntos
Citocromo P-450 CYP1A1/genética , Mercúrio/toxicidade , Transcrição Gênica/efeitos dos fármacos , Catálise , Sobrevivência Celular/efeitos dos fármacos , Citocromo P-450 CYP1A1/análise , Citocromo P-450 CYP1A1/metabolismo , Relação Dose-Resposta a Droga , Heme/farmacologia , Heme Oxigenase-1/genética , Células Hep G2 , Humanos , Dibenzodioxinas Policloradas/toxicidade , Processamento de Proteína Pós-Traducional , RNA Mensageiro/análise
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa