Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Pharmacol ; 65(5): 1302-12, 2004 May.
Article in English | MEDLINE | ID: mdl-15102959

ABSTRACT

The effects of inhibitors of 2,3-oxidosqualene:lanosterol cyclase (cyclase) on cytochrome P450 expression were investigated in primary cultures of rat hepatocytes. Treatment of hepatocyte cultures for 24 h with either of the inhibitors [4'-(6-allyl-methyl-amino-hexyloxy)-2'-fluoro-phenyl]-(4-bromophenyl)-methanone fumarate (Ro 48-8071) or trans-N-(4-chlorobenzoyl)-N-methyl-(4-dimethylaminomethylphenyl)-cyclohexylamine (BIBX 79) selectively increased CYP3A mRNA and immunoreactive protein contents, with maximal accumulations occurring at 3 x 10(-5) M Ro 48-8071 and 10(-4) M BIBX 79. The abilities of Ro 48-8071, BIBX 79, and 3beta-(2-diethylaminoethoxy)androst-5-en-17-one.HCl (U18666A) to induce murine CYP3A were abolished in hepatocyte cultures prepared from pregnane X receptor (PXR)-null mice, and cotransfection of primary cultured rat hepatocytes with a dominant-negative PXR prevented cyclase inhibitor-inducible luciferase expression from a PXR-responsive reporter plasmid. Cyclase inhibitor-mediated CYP3A mRNA induction was eliminated when primary cultured rat hepatocytes were cotreated with any of the following agents that inhibit steps upstream of cyclase in the cholesterol biosynthetic pathway: squalestatin 1 (squalene synthase inhibitor), (E)N-ethyl-N-(6,6-dimethyl-2-hepten-4-ynyl)-3-[(3,3'-bithiophen-5-yl)methoxy]benzenemethanamine (NB-598, squalene monooxygenase inhibitor), or pravastatin (HMG-CoA reductase inhibitor). Ro 48-8071-inducible CYP3A mRNA expression was restored when pravastatin-treated cultures were incubated with medium containing mevalonate. The concentration-dependence of Ro 48-8071-mediated CYP3A mRNA induction corresponded to the cellular contents of metabolically labeled squalene 2,3-oxide and squalene 2,3:22,23-dioxide, but not 24(S),25-epoxycholesterol. These results indicate that cyclase inhibitors are capable of inducing CYP3A expression in primary cultured rat and mouse hepatocytes and that the effect is mediated as a consequence of cyclase blockade through the evoked accumulation of one or more squalene metabolites that activate the PXR.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Benzophenones/pharmacology , Hepatocytes/drug effects , Intramolecular Transferases/antagonists & inhibitors , Oxidoreductases, N-Demethylating/biosynthesis , Squalene/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cells, Cultured , Cytochrome P-450 CYP3A , Enzyme Induction/drug effects , Hepatocytes/enzymology , Male , Mice , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley , Transfection
2.
Drug Metab Dispos ; 32(1): 66-71, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14709622

ABSTRACT

The effects of oxysterol and drug ligands of the liver X receptor (LXR) on cytochrome P450 expression were evaluated in primary cultured rodent hepatocytes. Treatment of rat hepatocyte cultures with either 25-hydroxycholesterol or 24(S),25-epoxycholesterol (10(-5) to 5 x 10(-5) M) produced concentration-dependent elevations in CYP3A mRNA and immunoreactive protein levels but did not increase the amounts of CYP1A1, CYP2B, or CYP4A gene products. The effects of 24(S),25-epoxycholesterol on CYP3A content were much greater than were those of 25-hydroxycholesterol, consistent with the relative abilities of these sterols to bind and activate LXR. To understand the mechanistic basis of these observations, experiments were performed using primary cultured hepatocytes prepared from LXRalpha/beta- or pregnane X receptor (PXR)-null mice. CYP3A mRNA levels were increased after treatment with 24(S),25-epoxycholesterol in both wild-type and LXR-null mouse hepatocytes. In contrast, neither 24(S),25-epoxycholesterol nor either of two additional potent LXR ligands, 22(R)-hydroxycholesterol and N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1(trifluoromethyl)ethyl-]phenyl]-benzenesulfonamide (T0901317), altered CYP3A mRNA levels in hepatocytes prepared from PXR-null mice, although these agents induced CYP3A mRNA content in wild-type cultures. As evidence that the LXR ligands also activated PXR in rat hepatocytes, cotransfection of primary cultures with a dominant negative PXR abolished reporter gene induction after treatment with any of the test agents. These results indicate that selected LXR ligands are capable of activating PXR, probably as a defensive measure to prevent the accumulation of these potentially toxic endogenous molecules.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Hepatocytes/enzymology , Oxidoreductases, N-Demethylating/biosynthesis , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Steroid/drug effects , Animals , Anticholesteremic Agents/pharmacology , Blotting, Northern , Blotting, Western , Cells, Cultured , Cytochrome P-450 CYP3A , DNA-Binding Proteins , Enzyme Induction/drug effects , Gene Expression Regulation/drug effects , Genes, Reporter/genetics , Hepatocytes/drug effects , Hydroxycholesterols/metabolism , Ligands , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors , Pregnane X Receptor , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Transfection
3.
Drug Metab Dispos ; 30(12): 1400-5, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12433810

ABSTRACT

The effects of treatment with the 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase) inhibitors lovastatin, simvastatin, pravastatin, fluvastatin, and atorvastatin on the contents of cytochrome p450 mRNAs were examined in primary cultures of human hepatocytes prepared from three different livers. Treatment of 2- to 3-day-old human hepatocyte cultures with 3 x 10(-5) M lovastatin, simvastatin, fluvastatin, or atorvastatin for 24 h increased the amounts of CYP2B6 and CYP3A mRNA by an average of 3.8- to 9.2-fold and 24- to 36-fold, respectively. In contrast, pravastatin treatment had no effect on the mRNA level of either CYP2B6 or CYP3A, although treatment with pravastatin did produce the expected compensatory increase in HMG-CoA reductase mRNA content, indicating effective inhibition of cholesterol biosynthesis. Although treatment with the active (+), but not the inactive (-), enantiomer of atorvastatin increased the amount of HMG-CoA reductase mRNA, treatment with each enantiomer significantly induced both CYP2B6 and CYP3A mRNA levels. Treatment of primary cultured rat hepatocytes with the atorvastatin enantiomers effectively increased the amount of CYP3A mRNA, but had no effect on CYP2B or CYP4A mRNA levels, in contrast to fluvastatin, which increased both. Findings for p450 proteins by Western blotting were consistent with the mRNA results. These findings indicate that the ability of a drug to inhibit HMG-CoA reductase activity does not predict its ability to produce p450 induction in primary cultured human hepatocytes, and demonstrate that some, but not all, of the effects of these drugs that occur in primary cultured rat hepatocytes are conserved in human hepatocyte cultures.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Hepatocytes/enzymology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Oxidoreductases, N-Demethylating/biosynthesis , Animals , Cells, Cultured , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Species Specificity
4.
Mol Pharmacol ; 62(5): 1177-86, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12391282

ABSTRACT

Because our previous studies indicated that squalestatin 1 treatment induces CYP2B expression in primary cultures of rat hepatocytes as a direct consequence of squalene synthase inhibition, we investigated possible underlying mechanisms. Cotransfection of cultured Sprague-Dawley male rat hepatocytes with each of the three sterol regulatory element binding protein (SREBP) transcription factors failed to induce luciferase expression from a squalestatin 1-responsive CYP2B1 reporter plasmid. Squalestatin 1 treatment of primary hepatocyte cultures from male Wistar-Kyoto rats produced a greater induction of CYP2B mRNA than occurred in cultures from female rats, consistent with the previously demonstrated response dimorphism that has been attributed to differences in constitutive androstane receptor (CAR) levels. Cotransfection of female Wistar-Kyoto rat hepatocyte cultures with plasmid expressing either mouse or rat CAR restored squalestatin 1-inducible CYP2B1-reporter expression. Cotransfection of Sprague-Dawley rat hepatocyte cultures with plasmid expressing rat CAR lacking the C-terminal AF-2 subdomain inhibited squalestatin 1-inducible CYP2B1-reporter expression. Squalestatin 1-mediated CYP2B mRNA induction in rat hepatocyte cultures was completely abolished by pretreatment with the 3-hydroxymethyl-3-glutaryl CoA reductase inhibitor pravastatin and was rescued by mevalonate supplementation, whereas phenobarbital-mediated induction was unaffected by these treatments. Finally, direct addition of trans,trans-farnesol to the culture medium caused the rapid induction of CYP2B mRNA. These results indicate that squalestatin 1 treatment induces CYP2B expression, not by inhibiting sterol synthesis and activating SREBPs, but by evoking the accumulation of an endogenous isoprenoid and activating CAR.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cytochrome P-450 CYP2B1/biosynthesis , Gene Expression/drug effects , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Tricarboxylic Acids/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Cells, Cultured , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B1/metabolism , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Hepatocytes/enzymology , Male , Rats , Rats, Sprague-Dawley
5.
J Pharmacol Toxicol Methods ; 47(3): 177-87, 2002.
Article in English | MEDLINE | ID: mdl-12628309

ABSTRACT

INTRODUCTION: To determine the feasibility of using dominant negative nuclear receptors to dissect the regulation of inducible gene expression in primary cultured hepatocytes, a series of dominant negative nuclear receptor expression plasmids were designed with truncated AF-2 subdomains. METHODS: Plasmids expressing dominant negative or wild-type constitutive androstane receptor (CAR), pregnane X receptor (PXR), farnesoid X receptor (FXR), liver X receptor (LXR), or peroxisome proliferator-activated receptor alpha (PPARalpha) were transiently cotransfected into primary cultured rat hepatocytes, together with an appropriate reporter plasmid. RESULTS: Treatment with prototypic inducers, 10(-4) M phenobarbital (CAR activator), 10(-5) M pregnenolone 16alpha-carbonitrile (PXR activator), 3x10(-5) M chenodeoxycholate (FXR activator), or 10(-4) M ciprofibrate (PPARalpha activator), significantly activated expression from the corresponding reporter plasmid. Treatment with 22(R)-hydroxycholesterol (LXR activator) only weakly activated the LXR-responsive reporter, while pregnenolone 16alpha-carbonitrile treatment significantly activated this reporter. Cotransfection with wild-type LXRalpha strongly enhanced 22(R)-hydroxycholesterol-inducible expression from the LXR-responsive reporter. Cotransfection of hepatocyte cultures with each of the dominant negative nuclear receptor plasmids significantly inhibited inducible expression of the corresponding reporter while, with one exception (LXRalpha), cotransfection with the wild-type receptor moderately enhanced or had little effect on reporter expression. When each dominant negative nuclear receptor was cross-examined against all inducer-reporter pairs, effects on multiple inducer-reporter pairs were frequently observed. However, in general, only cotransfection with the appropriate dominant negative inhibited inducible reporter expression to a greater extent than did cotransfection with the corresponding wild-type receptor. DISCUSSION: We suggest that the application of dominant negative nuclear receptors has utility in transient transfection studies aimed at discerning the regulatory role of individual nuclear receptor transcription factors in inducible hepatic gene expression, provided that appropriate controls are employed.


Subject(s)
Clofibric Acid/analogs & derivatives , Gene Expression/drug effects , Genes, Dominant , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Xenobiotics/pharmacology , Animals , Cells, Cultured , Chenodeoxycholic Acid/pharmacology , Clofibric Acid/pharmacology , Constitutive Androstane Receptor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibric Acids , Hepatocytes/metabolism , Male , Phenobarbital/pharmacology , Pregnane X Receptor , Pregnenolone Carbonitrile/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...