Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neuroscience ; 281: 124-34, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25280786

ABSTRACT

P450 metabolic enzymes are expressed in the human and rodent brain. Recent data support their involvement in the pathophysiology of epilepsy. However, the determinants of metabolic enzyme expression in the epileptic brain are unclear. We tested the hypothesis that status epilepticus (SE) or exposure to phenytoin or phenobarbital affects brain expression of the metabolic enzyme CYP2E1. SE was induced in C57BL/6J mice by systemic kainic acid. Brain CYP2E1 expression was evaluated 18-24h after severe SE by immunohistochemistry. Co-localization with neuronal nuclei (NEUN), glial fibrillary acidic protein (GFAP) and CD31 was determined by confocal microscopy. The effect of phenytoin, carbamazepine and phenobarbital on CYP2E1 expression was evaluated in vivo or by using organotypic hippocampal cultures in vitro. CYP2E1 expression was investigated in brain resections from a cohort of drug-resistant epileptic brain resections and human endothelial cultures (EPI-EC). Immunohistochemistry showed an increase of CYP2E1 expression limited to hippocampal CA2/3 and hilar neurons after severe SE in mice. CYP2E1 expression was also observed at the astrocyte-vascular interface. Analysis of human brain specimens revealed CYP2E1 expression in neurons and vascular endothelial cells (EC). CYP2E1 was expressed in cultured human EC and over-expressed by EPI-EC. When analyzing the effect of drug exposure on CYP2E1 expression we found that, in vivo or in vitro, ethanol increased CYP2E1 levels in the brain and liver. Treatment with phenytoin induced localized CYP2E1 expression in the brain whereas no significant effects were exerted by carbamazepine or phenobarbital. Our data indicate that the effect of acute SE on brain CYP2E1 expression is localized and cell specific. Exposure to selected anti-epileptic drugs could play a role in determining CYP2E1 brain expression. Additional investigation is required to fully reproduce the culprits of P450 enzyme expression as observed in the human epileptic brain.


Subject(s)
Anticonvulsants/pharmacology , Brain/metabolism , Central Nervous System Depressants/pharmacology , Cytochrome P-450 CYP2E1/metabolism , Endothelial Cells/metabolism , Ethanol/pharmacology , Neurons/metabolism , Phenytoin/pharmacology , Status Epilepticus/metabolism , Adolescent , Adult , Animals , Brain/drug effects , Carbamazepine/pharmacology , Cells, Cultured , Child, Preschool , Cytochrome P-450 CYP2E1/drug effects , Disease Models, Animal , Female , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenobarbital/pharmacology
2.
Gastroenterology ; 127(5): 1436-45, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15521013

ABSTRACT

BACKGROUND AND AIMS: Cytochrome P450 1A1 catalyzes the degradation of endobiotics (estradiol, fatty acids, and so on) and the bioactivation of numerous environmental procarcinogens, such as arylamines and polycyclic aromatic hydrocarbons, that are found in food. Several peroxisome proliferators and arachidonic acid derivatives enhance cytochrome P450 1A1 activity, but the mechanisms involved remain unknown. The aim of this work was to study the role of peroxisome proliferator-activated receptors in cytochrome P450 1A1 gene induction. METHODS: The role of peroxisome proliferator-activated receptor transcription factors in cytochrome P450 1A1 induction was assessed by means of enzymatic activities, quantitative real-time polymerase chain reaction, gene reporter assays, mutagenesis, and electrophoretic mobility shift assay. RESULTS: We show that peroxisome proliferator-activated receptor-alpha agonists (WY-14643, bezafibrate, clofibrate, and phthalate) induce human cytochrome P450 1A1 gene expression, whereas 2,4-thiazolidinedione, a specific peroxisome proliferator-activated receptor-gamma agonist, represses it. The induction of cytochrome P450 1A1 transcripts by WY-14643 was associated with a marked increase of ethoxyresorufin O -deethylase activity (10-fold at 200 mumol/L). Transfection of peroxisome proliferator-activated receptor-alpha complementary DNA enhanced cytochrome P450 1A1 messenger RNA induction by WY-14643, although WY-14643 failed to activate xenobiotic responsive element sequences. Two peroxisome proliferator response element sites were located at positions -931/-919 and -531/-519 of the cytochrome P450 1A1 promoter. Their inactivation by directed mutagenesis suppressed the inductive effect of WY-14643 on cytochrome P450 1A1 promoter activation. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay experiments showed that the 2 cytochrome P450 1A1 peroxisome proliferator response element sites bind the peroxisome proliferator-activated receptor-alpha/retinoid X receptor-alpha heterodimer. CONCLUSIONS: We describe here a new cytochrome P450 1A1 induction pathway involving peroxisome proliferator-activated receptor-alpha and 2 peroxisome proliferator response element sites, indicating that peroxisome proliferator-activated receptor-alpha ligands, which are common environmental compounds, may be involved in carcinogenesis.


Subject(s)
Cytochrome P-450 CYP1A1/genetics , PPAR alpha/physiology , Adenocarcinoma , Base Sequence , Carcinoma, Hepatocellular , Cell Line, Tumor , Chloramphenicol O-Acetyltransferase/metabolism , Colonic Neoplasms , DNA Primers , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms , PPAR alpha/genetics , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , RNA, Messenger/genetics , Transcriptional Activation
3.
Xenobiotica ; 34(7): 633-64, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15672753

ABSTRACT

1. Recent investigations on nuclear receptors and other transcription factors involved in the regulation of genes encoding xenobiotic metabolizing and transport systems reveal that xenobiotic-dependent signalling pathways are embedded in, and establish functional interactions with, a tangle of regulatory networks involving the glucocorticoid and oestrogen receptors, the hypoxia-inducible factor, the vitamin D receptor and other transcription factors/nuclear receptors controlling cholesterol/bile salt homeostasis and liver differentiation. 2. Such functional interferences provide new insight, first for understanding how xenobiotics might exert adverse effects, and second how physiopathological stimuli affect xenobiotic metabolism.


Subject(s)
Inactivation, Metabolic/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcription Factors/metabolism , Xenobiotics/metabolism , Animals , Humans , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Transcription, Genetic
4.
Biochim Biophys Acta ; 1619(3): 243-53, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12573484

ABSTRACT

Numerous chemicals increase the metabolic capability of organisms by their ability to activate genes encoding various xenochemical-metabolizing enzymes, such as cytochromes P450 (CYPs), transferases and transporters. For example, natural and synthetic glucocorticoids (agonists and antagonists) as well as other clinically important drugs induce the hepatic CYP2B, CYP2C and CYP3A subfamilies in man, and these inductions might lead to clinically important drug-drug interactions. Only recently, the key cellular receptors that mediate such inductions have been identified. They include nuclear receptors, such as the constitutive androstane receptor (CAR, NR1I3), the retinoid X receptor (RXR, NR2B1), the pregnane X receptor (PXR, NR1I2), and the vitamin D receptor (VDR, NR1I1) and steroid receptors such as the glucocorticoid receptor (GR, NR3C1). There is a wide promiscuity of these receptors in the induction of CYPs in response to xenobiotics. Indeed, this adaptive system appears now as a tangle of networks, where receptors share partners, ligands, DNA response elements and target genes. Moreover, they influence mutually their relative expression. This review is focused on these different pathways controlling human CYP2B6, CYP2C9 and CYP3A4 gene expression, and the cross-talk between these pathways.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 Enzyme System/genetics , Oxidoreductases, N-Demethylating/genetics , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Colon/metabolism , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , Gene Expression Regulation, Enzymologic , Glucocorticoids/pharmacology , Humans , Intestine, Small/metabolism , Liver/metabolism , Oxidoreductases, N-Demethylating/biosynthesis , Pregnane X Receptor , Receptors, Calcitriol/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/metabolism , Species Specificity , Transcription Factors/metabolism , Xenobiotics/pharmacology
5.
Eur J Biochem ; 268(24): 6346-58, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11737189

ABSTRACT

Although CYP3A induction by dexamethasone has been extensively documented, its mechanism is still unclear because both the role of the glucocorticoid receptor and the ability of dexamethasone to activate the human pregnane X receptor have been questioned. In an attempt to resolve this problem, we investigated the response of CYP3A4 to dexamethasone (10 nm-100 microm) in primary human hepatocytes and HepG2 cells, using a variety of methods: kinetic analysis of CYP3A4 and tyrosine aminotransferase expression, effects of RU486 and cycloheximide, ligand binding assay, cotransfection of HepG2 cells with CYP3A4 reporter gene constructs and vectors expressing the glucocorticoid receptor, pregnane X receptor or constitutively activated receptor. In contrast to rifampicin (monophasic induction), dexamethasone produces a biphasic induction of CYP3A4 mRNA consisting of a low-dexamethasone component (nmol concentrations) of low amplitude (factor of 3-4) followed by a high-dexamethasone component (supramicromolar concentrations) of high amplitude (factor of 15-30). We show that the low-dexamethasone component results from the glucocorticoid receptor-mediated expression of pregnane X receptor and/or constitutively activated receptor which, in turn, are able to transactivate CYP3A4 in a xenobiotic-independent manner. At supramicromolar concentrations (>10 microm), dexamethasone binds to and activates pregnane X receptor thus producing the high-dexamethasone component of CYP3A4 induction. We conclude that, in contrast to the other xenobiotic inducers of CYP3A4, glucocorticoids play a dual role in CYP3A4 expression, first by controlling the expression of PXR and CAR under physiological conditions (submicromolar concentrations) through the classical glucocorticoid receptor pathway, and second by activating the pregnane X receptor under bolus or stress conditions (supramicromolar concentrations).


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Dexamethasone/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Mixed Function Oxygenases/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Glucocorticoid/physiology , Receptors, Steroid/physiology , Animals , Base Sequence , Cell Line , Cycloheximide/pharmacology , Cytochrome P-450 CYP3A , DNA Primers , Hepatocytes/enzymology , Humans , Pregnane X Receptor , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
Drug Metab Dispos ; 29(10): 1325-31, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11560876

ABSTRACT

The expression of three cytochromes P450 (CYP3A4, CYP2C9, and CYP2B6) was investigated in primary human hepatocyte cultures following treatment with four calcium channel modulators (CCM) of the dihydropyridine family, three antagonists (nifedipine, nicardipine, and isradipine), and one agonist (BK8644). Induction of CYP3A4 was studied by Northern blot, Western blot, and enzymatic activity. Induction began between 1 and 10 microM CCM and was dependent on the presence of dexamethasone (100 nM) in the medium. CYP3A4 mRNA accumulation started only after 16 h of treatment because pregnane X receptor (hPXR) synthesis was needed. Cotransfection experiments showed that the proximal and the distal PXR response elements of the CYP3A4 promoter and hPXR (HepG2 cells) or dexamethasone-induced hPXR (primary hepatocytes) were necessary to obtain full induction. Furthermore, glutathione S-transferase pull-down assays demonstrated that the CCM tested can act as hPXR ligands. In addition, cotransfection experiments in CV1 cells showed that these compounds failed to reverse CAR (constitutively activated receptor) inactivation by androstenol. Finally, 10 microM CCM induced both CYP2C9 and CYP2B6, strengthening the evidence that hPXR is involved in the regulation of these genes. All together, these results widen the field of hPXR activators to a new class of ligand, namely the CCM of the dihydropyridine family.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Calcium Channels/metabolism , Cytochrome P-450 Enzyme System/biosynthesis , Dihydropyridines/pharmacology , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Steroid 16-alpha-Hydroxylase , Calcium Channels/drug effects , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Hepatocytes/enzymology , Humans , Kinetics , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/genetics , Nifedipine/pharmacology , Oxidoreductases, N-Demethylating/biosynthesis , Pregnane X Receptor , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rifampin/pharmacology , Steroid Hydroxylases/biosynthesis , Time Factors
7.
Drug Metab Dispos ; 29(3): 242-51, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181490

ABSTRACT

The expression and inducibility of four CYP2C genes, including CYP2C8, -2C9, -2C18, and -2C19, was investigated in primary cultures of human hepatocytes. By the use of RNase protection assay and specific antibodies, each CYP2C mRNA and protein were quantified unequivocally. The four CYP2C mRNAs were expressed in human livers and cultured primary hepatocytes, but only the CYP2C18 protein was not detected. Compounds known to activate the pregnane X receptor (PXR) such as rifampicin, or the constitutively activated receptor (CAR) such as phenobarbital, induced CYP2C8, CYP2C9, and to a lesser extent CYP2C19 mRNAs and proteins. CYP2C18 mRNA was expressed but not inducible. The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. In contrast, dexamethasone produced maximum induction of CYP2C8 and CYP2C9 mRNAs at 0.1 microM while in these conditions neither CYP3A4 nor CYP2B6 was significantly induced. Moreover, the concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to dexamethasone paralleled that of tyrosine aminotransferase. Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Collectively, these results suggest the possible implication of at least three receptors in the regulation of CYP2C8 and CYP2C9 expression, i.e., glucocorticoid receptor, PXR, and/or CAR.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Hepatocytes/enzymology , Adult , Aged , Cell Line , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dactinomycin/pharmacology , Dexamethasone/pharmacology , Enzyme Induction/drug effects , Female , Hepatocytes/metabolism , Humans , Hydroxylation , Immunoblotting , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Liver/enzymology , Liver/metabolism , Male , Middle Aged , Nuclease Protection Assays , Phenobarbital/pharmacology , Pregnane X Receptor , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/metabolism , Receptors, Virus/metabolism , Rifampin/pharmacology , Tyrosine Transaminase/genetics , Tyrosine Transaminase/metabolism
8.
Mol Pharmacol ; 58(6): 1441-50, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11093784

ABSTRACT

The barbiturate phenobarbital induces the transcription of cytochromes P450 (CYPs) 2B through the constitutive androstane receptor (CAR; NR1I3). CAR is a member of the nuclear receptor family (NR1) mostly expressed in the liver, which heterodimerizes with retinoid X receptor (RXR) and was shown to transactivate both the phenobarbital responsive element module of the human CYP2B6 gene and the CYP3A4 xenobiotic response element. Because previous studies in rodent hepatocyte cultures have shown that the phenobarbital-mediated induction of CYP2B genes is potentiated by glucocorticoids, we examined the role of activated glucocorticoid receptor in this process. We show that submicromolar concentrations of dexamethasone enhance phenobarbital-mediated induction of CYP3A4, CYP2B6, and CYP2C8 mRNA in cultured human hepatocytes. In parallel, we observed that glucocorticoid agonists, such as dexamethasone, prednisolone, or hydrocortisone, specifically increase human car (hCAR) mRNA expression. Accumulation of hCAR mRNA parallels that of tyrosine aminotransferase: both mRNAs reach a maximum at a concentration of 100 nM dexamethasone and are down-regulated by concomitant treatment with the glucocorticoid antagonist RU486. Moreover, the effect of dexamethasone on hCAR mRNA accumulation appears to be of transcriptional origin because the addition of protein synthesis inhibitor cycloheximide has no effect, and dexamethasone does not affect the degradation of hCAR mRNA. Furthermore, dexamethasone increases both basal and phenobarbital-mediated nuclear translocation of CAR immunoreactive protein in human hepatocytes. The up-regulation of CAR mRNA and protein in response to dexamethasone explains the synergistic effect of this glucocorticoid on phenobarbital-mediated induction of CYP2B genes and the controversial role of the glucocorticoid receptor on phenobarbital-mediated CYP gene inductions.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Dexamethasone/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/biosynthesis , Steroid 16-alpha-Hydroxylase , Transcription Factors/biosynthesis , Active Transport, Cell Nucleus/drug effects , Cells, Cultured , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Hepatocytes/physiology , Humans , Liver/metabolism , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/genetics , Oxidoreductases, N-Demethylating/biosynthesis , Oxidoreductases, N-Demethylating/genetics , Phenobarbital/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Glucocorticoid/physiology , Steroid Hydroxylases/biosynthesis , Steroid Hydroxylases/genetics , Transcription Factors/genetics , Transcriptional Activation
9.
Biochem Biophys Res Commun ; 274(3): 707-13, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10924340

ABSTRACT

The marked impairment of hepatic drug metabolism during inflammation and infections has been known for many years and shown to result from down-regulation of cytochrome P450s (CYP) by cytokines. However, the mechanism of this repression is unknown. Using primary cultures of human hepatocytes, we show here that interleukin-6 (IL-6) rapidly and markedly decreases the expression of PXR (pregnane X receptor) and CAR (constitutively activated receptor) mRNAs, but does not affect the levels of dioxin receptor and glucocorticoid receptor mRNA. In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. As the transcriptional activity of PXR and CAR is not affected by IL-6 in cell-based reporter assays, our data suggest that the loss of CYP2 and CYP3 inducibility results from the negative regulation of PXR and CAR gene expression by this cytokine.


Subject(s)
Interleukin-6/pharmacology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription Factors , Cells, Cultured , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Down-Regulation , Humans , Interleukin-6/metabolism , Pregnane X Receptor
10.
Mol Pharmacol ; 58(2): 361-72, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10908304

ABSTRACT

In this report we show that submicromolar concentrations of dexamethasone enhance pregnane X receptor (PXR) activator-mediated CYP3A4 gene expression in cultured human hepatocytes. Because this result is only observed after 24 h of cotreatment and is inhibited by pretreatment with cycloheximide, we further investigated which factor(s), induced by dexamethasone, might be responsible for this effect. We report that dexamethasone increases both retinoid X receptor-alpha (RXRalpha) and PXR mRNA expression in cultured human hepatocytes, whereas PXR activators such as rifampicin and clotrimazole do not. Accumulation of RXRalpha and PXR mRNA reaches a maximum at a concentration of 100 nM dexamethasone after treatment for 6 to 12 h and is greatly diminished by RU486. A similar pattern of expression is observed with tyrosine aminotransferase mRNA. Moreover, the effect of dexamethasone on PXR mRNA accumulation seems to be through direct action on the glucocorticoid receptor (GR) because the addition of cycloheximide has no effect, and dexamethasone does not affect the degradation of PXR mRNA. Furthermore, dexamethasone induces the accumulation of a RXRalpha-immunoreactive protein and increases the nuclear level of RXRalpha:PXR heterodimer as shown by gel shift assays with a CYP3A4 ER6 PXRE probe. This accumulation of latent PXR and RXRalpha in the nucleus of hepatocytes explains the synergistic effect observed with dexamethasone and PXR activators together on CYP3A4 induction. These results reveal the existence of functional cross talk between the GR and PXR, and may explain some controversial aspects of the role of the GR in CYP3A4 induction.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Dexamethasone/pharmacology , Liver/drug effects , Mixed Function Oxygenases/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Receptors, Steroid/biosynthesis , Transcription Factors/biosynthesis , Adult , Aged , Biological Transport , Cell Extracts , Cells, Cultured , Cytochrome P-450 CYP3A , Enzyme Induction , Female , Gene Expression/drug effects , Glucocorticoids/pharmacology , Humans , Liver/cytology , Liver/metabolism , Male , Middle Aged , Nucleic Acid Conformation , Pregnane X Receptor , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Steroid/agonists , Receptors, Steroid/genetics , Retinoid X Receptors , Signal Transduction , Transcription Factors/genetics
11.
Biochem Biophys Res Commun ; 260(2): 377-81, 1999 Jul 05.
Article in English | MEDLINE | ID: mdl-10403778

ABSTRACT

Pregnane X Receptor (PXR) has been recently shown to regulate the inducible expression of CYP3A genes in response to xenobiotics and steroids. PXR forms a heterodimer with the retinoic acid receptor (RXR) and this complex binds to and transactivates an 18bp region containing two everted repeats TGA(A/C)CT separated by 6 nucleotides (ER6) and located at approximately -150 in the CYP3A4 promoter. In this work we have isolated and sequenced the proximal 5'-flanking region of CYP3A7 from two different human genomic libraries. In contrast to a previously reported sequence (Itoh et al., 1992), we did not observe any mutation in the 3'-half of the CYP3A7 ER6 element. Using electrophoretic mobility shift assays and cotransfection experiments we show that this element is able to bind the PXR:RXR complex and transactivates the expression of a down stream promoter in response to rifampicin, clotrimazole, and RU-486, three compounds known to specifically activate the human PXR. This is consistent with the fact that CYP3A7 mRNA is inducible in several primary cultures of human hepatocytes from different patients, as well as in two hepatocarcinoma cell lines HuH7 and HepG2, in response to these compounds. In contrast to a previous report (Blumberg et al., 1998), based on the sequence published by Itoh et al., we conclude that CYP3A7, like CYP3A4, is inducible in response to xenobiotics and presumably in a large proportion of the population.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Adult , Base Sequence , Cell Line , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , DNA Primers , Humans , Pregnane X Receptor , Protein Binding , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Transcriptional Activation
12.
J Biol Chem ; 274(20): 14229-37, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318843

ABSTRACT

T-cell antigen receptor-induced signaling requires both ZAP-70 and Lck protein-tyrosine kinases. One essential function of Lck in this process is to phosphorylate ZAP-70 and up-regulate its catalytic activity. We have previously shown that after T-cell antigen receptor stimulation, Lck binds to ZAP-70 via its Src homology 2 (SH2) domain (LckSH2) and, more recently, that Tyr319 of ZAP-70 is phosphorylated in vivo and plays a positive regulatory role. Here, we investigated the possibility that Tyr319 mediates the SH2-dependent interaction between Lck and ZAP-70. We show that a phosphopeptide encompassing the motif harboring Tyr319, YSDP, interacted with LckSH2, although with a lower affinity compared with a phosphopeptide containing the optimal binding motif, YEEI. Moreover, mutation of Tyr319 to phenylalanine prevented the interaction of ZAP-70 with LckSH2. Based on these results, a gain-of-function mutant of ZAP-70 was generated by changing the sequence Y319SDP into Y319EEI. As a result of its increased ability to bind LckSH2, this mutant induced a dramatic increase in NFAT activity in Jurkat T-cells, was hyperphosphorylated, and displayed a higher catalytic activity compared with wild-type ZAP-70. Collectively, our findings indicate that Tyr319-mediated binding of the SH2 domain of Lck is crucial for ZAP-70 activation and consequently for the propagation of the signaling cascade leading to T-cell activation.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , Tyrosine/metabolism , src Homology Domains , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Catalysis , Humans , Jurkat Cells , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenotype , Phosphorylation , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, T-Cell/genetics , Structure-Activity Relationship , Up-Regulation , ZAP-70 Protein-Tyrosine Kinase
13.
Nat Med ; 4(1): 92-6, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9427613

ABSTRACT

The glucocorticoid receptor (GR) belongs to a superfamily of ligand-regulated nuclear steroid hormone receptors. The steps in the signal transduction pathway leading to the biological effects of glucocorticoids (GCs) include sequentially binding of the steroid to the GR ligand binding domain (LBD), receptor transformation, nuclear translocation and either positive or negative gene transactivation. Rifampicin (RIF) is a macrocyclic antibiotic used as an antituberculosis agents. As the incidence of tuberculosis has been increasing, in part because of the AIDS epidemic, a growing number of patients are being exposed to the adverse effects of this antibiotic. Indeed, this compound, as are the GCs, is often implicated in noxious drug interactions, because of its strong ability to induce drug-metabolizing enzymes. Moreover, in humans, RIF, as are the GCs, has been described as a potential immunodepressor, associated notably with the reduction of mitogenic responsiveness of human peripheral blood lymphocytes. Here, we report that RIF activates the human glucocorticoid receptor (hGR). Transient expression of wild-type, deleted or mutated GRs; sucrose density gradient sedimentation; and the BIAcore technique strongly suggest that RIF binds to the receptor with the physiological consequence that this antibiotic acts as an immunodepressor. Given the wide use of RIF in the treatment of coinfection of tuberculosis and HIV, this report is highly relevant to current medical practice.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Rifampin/pharmacology , Animals , Antibiotics, Antitubercular/metabolism , CHO Cells , COS Cells , Cricetinae , Dexamethasone/metabolism , Dexamethasone/pharmacology , Humans , Kinetics , Ligands , Luciferases/biosynthesis , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Rifampin/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...