Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Pharmacol ; 69(5): 1554-63, 2006 May.
Article in English | MEDLINE | ID: mdl-16449384

ABSTRACT

trans-Stilbene oxide (TSO) induces drug metabolizing enzymes in rat and mouse liver. TSO is considered a phenobarbital-like compound because it induces Cyp2B mRNA expression in liver. Phenobarbital increases Cyp2B expression in liver via activation of the constitutive androstane receptor (CAR). The purpose of this study was to determine whether TSO induces gene expression in mouse liver via CAR activation. TSO increased CAR nuclear localization in mouse liver, activated the human Cyp2B6 promoter in liver in vivo, and activated a reporter plasmid that contains five nuclear receptor 1 (NR1) binding sites in HepG2 cells. TSO administration increased expression of Cyp2b10, NAD(P)H:quinone oxidoreductase (Nqo1), epoxide hydrolase, heme oxygenase-1, UDP-glucuronosyl-transferase (Ugt) 1a6 and 2b5, and multidrug resistance-associated proteins (Mrp) 2 and 3 mRNA in livers from male mice. Cyp2b10 and epoxide hydrolase induction by TSO was decreased in livers from CAR-null mice, compared with wild-type mice, suggesting CAR involvement. In contrast, TSO administration induced Nqo1 and Mrp3 mRNA expression equally in livers from wild-type and CAR-null mice, suggesting that TSO induces expression of some genes through a mechanism independent of CAR. TSO increased nuclear staining of the transcription factor Nrf2 in liver, and activated an antioxidant/electrophile response element luciferase reporter construct that was transfected into HepG2 cells. In summary, in mice, TSO increases Cyp2b10 and epoxide hydrolase expression in mice via CAR, and potentially induces Nqo1 and Mrp3 expression via Nrf2. Moreover, our data demonstrate that a single compound can activate both CAR and Nrf2 transcription factors in liver.


Subject(s)
Gene Expression Regulation/drug effects , Liver/physiology , NF-E2-Related Factor 2/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stilbenes/pharmacology , Transcription Factors/metabolism , Animals , Base Sequence , Cell Nucleus/metabolism , Constitutive Androstane Receptor , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Multidrug Resistance-Associated Protein 2 , Oligonucleotide Probes
2.
Biochem Pharmacol ; 61(2): 215-25, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11163336

ABSTRACT

To investigate a possible role of phosphorylation in the signal transduction pathways responsible for transcriptional regulation of drug-metabolizing enzymes, we tested seven specific tyrosine kinase inhibitors (tyrphostins) for their effects on NAD(P)H:quinone oxidoreductase-1 (NQO1) mRNA levels in mouse hepatoma Hepa-1c1c7 (Hepa-1) cells and chose to study AG879 further. The potent electrophile tert-butylhydroquinone (tBHQ) is known to activate NQO1 gene transcription via the electrophile response element (EPRE). Among the tyrphostins tested, tyrphostin AG879 was unique in preventing the accumulation of tBHQ-induced NQO1 mRNA; this effect was dependent on the AG879 dose and was also sensitive to the time when AG879 was added relative to the beginning of tBHQ treatment. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (dioxin; TCDD) is known to activate Cyp1a1 gene transcription by way of aromatic hydrocarbon response elements (AHREs). We found that AG879 also prevents, to a lesser extent, the AHRE-mediated induction of CYP1A1 and NQO1 mRNA by dioxin. Zinc or cadmium is known to activate metallothionein (Mt1) gene transcription via the metal response element (MRE). AG879 induced MT1 mRNA, and AG879 did not block zinc- or cadmium-induced MT1 mRNA, indicating that the effects of AG879 on NQO1 or CYP1A1 mRNA levels cannot be generalized to all transcripts. Using transient transfection of EPRE-, AHRE-, or MRE-driven luciferase reporter gene constructs in Hepa-1 cells, we showed that the inhibitory effects of AG879 occurred at the level of EPRE- and AHRE-mediated transcription, but that AG879 did not affect the MRE-driven transcriptional response. These data suggest that AG879 might inhibit an unknown tyrosine kinase(s) whose activity is essential for EPRE- and AHRE-mediated trans-activation of certain mammalian genes. These results also indicate that some sharing of common signal transduction pathways might exist in the regulation of genes involved in drug metabolism that also respond to oxidative stress.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Transcriptional Activation/drug effects , Tyrphostins/pharmacology , Animals , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Dioxins/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction , Hydroquinones/pharmacology , Metallothionein/biosynthesis , Metallothionein/genetics , Mice , Polychlorinated Dibenzodioxins/pharmacology , Quinone Reductases/biosynthesis , Quinone Reductases/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Time Factors , Tumor Cells, Cultured
3.
Cardiovasc Toxicol ; 1(4): 285-98, 2001.
Article in English | MEDLINE | ID: mdl-12213967

ABSTRACT

Epidemiologic studies have linked dioxin exposure to increased mortality caused by ischemic heart disease. To test the hypothesis that dioxin exposure may constitute an environmental risk factor for atherosclerosis, we exposed C57BL/6J mice to 5 microg/kg of dioxin daily for 3 d, and measured various molecular and physiological markers of heart disease. Dioxin treatment led to an increase in the urinary excretion of vasoactive eicosanoids and an elevation in the mean tail-cuff blood pressure. In addition, dioxin exposure led to an increase in triglycerides, but not in high-density lipoproteins, in both Apoe(+/+) mice and in hyperlipidemic Apoe(-/- mice. Dioxin exposure also led to an increase in low-density lipoproteins in Apoe(-/-) mice. After treatment, dioxin was associated with low-density lipoprotein particles, which might serve as a vehicle to deliver the compound to atherosclerotic plaques. Dioxin treatment of vascular smooth-muscle cells taken from C57Bl/6J mice resulted in the deregulation of several genes involved in cell proliferation and apoptosis. Subchronic treatment of Apoe(-/-) mice with dioxin (150 ng/kg, three times weekly) for 7 or 26 wk caused a trend toward earlier onset and greater severity of atherosclerotic lesions compared to those of vehicle treated mice. These results suggest that dioxin may increase the incidence of ischemic heart disease by exacerbating its severity.


Subject(s)
Environmental Pollutants/toxicity , Myocardial Ischemia/chemically induced , Polychlorinated Dibenzodioxins/toxicity , Animals , Aorta, Thoracic/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/chemically induced , Arteriosclerosis/pathology , Arteriosclerosis/physiopathology , Blood Pressure/drug effects , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cells, Cultured , Cholesterol/blood , Eicosanoids/metabolism , Lipoproteins/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/pathology , RNA/genetics , RNA/isolation & purification , Risk Factors
4.
Biochem Biophys Res Commun ; 279(2): 324-9, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11118286

ABSTRACT

The biosynthesis of reduced glutathione (GSH) is carried out by the enzymes gamma-glutamylcysteine synthetase (GCL) and GSH synthetase. GCL is the rate-limiting step and represents a heterodimeric enzyme comprised of a catalytic subunit (GCLC) and a ("regulatory"), or modifier, subunit (GCLM). The nonhomologous Gclc and Gclm genes are located on mouse chromosomes 9 and 3, respectively. GCLC owns the catalytic activity, whereas GCLM enhances the enzyme activity by lowering the K(m) for glutamate and increasing the K(i) to GSH inhibition. Humans have been identified with one or two defective GCLC alleles and show low GSH levels. As an initial first step toward understanding the role of GSH in cellular redox homeostasis, we have targeted a disruption of the mouse Gclc gene. The Gclc(-/-) homozygous knockout animal dies before gestational day 13, whereas the Gclc(+/-) heterozygote is viable and fertile. The Gclc(+/-) mouse exhibits a gene-dose decrease in the GCLC protein and GCL activity, but only about a 20% diminution in GSH levels and a compensatory increase of approximately 30% in ascorbate-as compared with that in Gclc(+/+) wild-type littermates. These data show a reciprocal action between falling GSH concentrations and rising ascorbate levels. Therefore, the Gclc(+/-) mouse may be a useful genetic model for mild endogenous oxidative stress.


Subject(s)
Chromosome Mapping , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/deficiency , Alleles , Animals , Catalytic Domain , Disease Models, Animal , Female , Genotype , Glutamate-Cysteine Ligase/deficiency , Heterozygote , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Polymerase Chain Reaction , Protein Subunits
5.
Pharmacology ; 61(3): 124-35, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10971198

ABSTRACT

So-called 'drug-metabolizing enzyme' (DME) genes have existed on this planet for more than 2.5 billion years and would be more appropriately named 'effector-metabolizing enzymes'. Genes encoding DMEs have functioned in many fundamental processes in prokaryotes and, more recently, in countless critical life processes in plants and animals. DME genes exist in every eukaryotic cell and in most, if not all, prokaryotes. Over the past decade, it has become clear that each person has their own 'individual fingerprint' of unique alleles coding for DMEs. The underlying genetic predisposition of each patient reflects combinations of poor- and extensive-metabolizer phenotypes. If these enzymes cooperate in the same metabolic pathway for any given drug or environmental agent, such ecogenetic variability might be synergistic and could cause 30- to > 40-fold differences in activation or degradation. The end result can be large interindividual differences in risk of environmentally caused toxicity or cancer. Human DME gene polymorphisms often show high frequencies of variant alleles. Many factors contribute to persistence of these high frequencies, including a combination of selective pressures involving diet, climate and geography, as well as 'balanced polymorphisms' ('shared benefit' for the heterozygote). However, the extensive heterogeneity in the human genome currently being discovered suggests many more polymorphisms will occur not only in drug metabolism genes, but in all genes, and exhibiting large gene-by-gene variability.


Subject(s)
Biological Evolution , Enzymes/metabolism , Pharmaceutical Preparations/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Enzymes/genetics , Humans , Plants , Polymorphism, Genetic/genetics
6.
Biochem Biophys Res Commun ; 267(1): 184-9, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10623596

ABSTRACT

Using the Cre-lox system, we have generated a cytochrome P450 1A1 Cyp1a1(-/-) knockout mouse by deletion of the translated portions of the Cyp1a1 gene. These mice are viable and demonstrate no obvious phenotype, compared with wild-type littermates. As a first step toward characterizing genes that might be expected to compensate for loss of CYP1A1, constitutive expression of [Ah] gene battery members was examined. In a cultured hepatoma CYP1A1 metabolism-deficient mutant line that does not express Cyp1a2, we have previously shown that constitutive transcriptional up-regulation of other [Ah] gene battery members occurs; these results are consistent with the elevation of a putative endogenous ligand (EL) for the Ah receptor that is a substrate for CYP1A1. The [Ah] battery includes Cyp1a2, NAD(P)H:quinone oxidoreductase (Nqo1), and three other Phase II genes. Examining mRNA, protein, and enzyme activity, we demonstrate that the absence of CYP1A1 has no effect on the hepatic constitutive expression of Cyp1a2 or Nqo1. We postulate that CYP1A1 and CYP1A2 might have overlapping substrate specificity for metabolism of the EL, such that basal CYP1A2 in the liver can compensate for the loss of CYP1A1.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP1A1/deficiency , Cytochrome P-450 CYP1A1/genetics , Gene Expression Regulation , Receptors, Aryl Hydrocarbon/genetics , Animals , Carcinoma, Hepatocellular , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 Enzyme System/genetics , Female , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxidoreductases/genetics , Protein Biosynthesis , Sequence Deletion , Tumor Cells, Cultured
7.
Biochem Pharmacol ; 59(1): 65-85, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10605936

ABSTRACT

The chronology and history of characterizing the aromatic hydrocarbon [Ah] battery is reviewed. This battery represents the Ah receptor (AHR)-mediated control of at least six, and probably many more, dioxin-inducible genes; two cytochrome P450 genes-P450 1A1 and 1A2 (Cypla1, Cypla2-and four non-P450 genes, have experimentally been documented to be members of this battery. Metabolism of endogenous and exogenous substrates by perhaps every P450 enzyme, but certainly CYP1A1 and CYP1A2 (which are located, in part, in the mitochondrion), have been shown to cause reactive oxygenated metabolite (ROM)-mediated oxidative stress. Oxidative stress activates genes via the electrophile response element (EPRE) DNA motif, whereas dioxin (acutely) activates genes via the AHR-mediated aromatic hydrocarbon response element (AHRE) DNA motif. In contrast to dioxin, AHR ligands that are readily metabolized to ROMs (e.g. benzo[a]pyrene, beta-naphthoflavone) activate genes via both AHREs and the EPRE. The importance of the AHR in cell cycle regulation and apoptosis has just begun to be realized. Current evidence suggests that the CYP1A1 and CYP1A2 enzymes might control the level of the putative endogenous ligand of the AHR, but that CYPA1/1A2 metabolism generates ROM-mediated oxidative stress which can be ameliorated by the four non-P450 EPRE-driven genes in the [Ah] battery. Oxidative stress is a major signal in precipitating apoptosis; however, the precise mechanism, or molecule, which determines the cell's decision between apoptosis and continuation with the cell cycle, remains to be elucidated. The total action of AHR and the [Ah] battery genes therefore represents a pivotal upstream event in the apoptosis cascade, providing an intricate balance between promoting and preventing ROM-mediated oxidative stress. These proposed endogenous functions of the AHR and [Ah] enzymes are, of course, in addition to the frequently described functions of "metabolic potentiation" and "detoxification" of various foreign chemicals.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , Oxidative Stress/physiology , Receptors, Aryl Hydrocarbon/physiology , Animals , Glutathione/metabolism , Humans , Receptor Cross-Talk , Signal Transduction , Tyrosine/metabolism , Tyrosinemias/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...