Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hepatology ; 49(6): 2068-79, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19437491

RESUMEN

The pregnane X receptor (PXR) initially isolated as a nuclear receptor regulating xenobiotic and drug metabolism and elimination, seems to play an endobiotic role by affecting lipid homeostasis. In mice, PXR affects lipid homeostasis and increases hepatic deposit of triglycerides. In this study, we show that, in human hepatocyte, PXR activation induces an increase of de novo lipogenesis through the up-regulation of S14. S14 was first identified as a thyroid-responsive gene and is known to transduce hormone-related and nutrient-related signals to genes involved in lipogenesis through a molecular mechanism not yet elucidated. We demonstrate that S14 is a novel transcriptional target of PXR. In addition, we report an increase of fatty acid synthase (FASN) and adenosine triphosphate citrate lyase genes expression after PXR activation in human hepatocyte, leading to an increase of fatty acids accumulation and de novo lipogenesis. RNA interference of the expression of S14 proportionally decreases the FASN induction, whereas S14 overexpression in human hepatic cells provokes an increase of fatty acids accumulation and lipogenesis. These results demonstrate for the first time that xenobiotic or drug-activated PXR promote aberrant hepatic de novo lipogenesis via activation of the nonclassical S14 pathway. In addition, these data suggest that the up-regulation of S14 by PXR may promote aberrant hepatic lipogenesis and hepatic steatosis in human hepatocytes.


Asunto(s)
Hepatocitos/metabolismo , Lipogénesis , Proteínas Nucleares/fisiología , Receptores de Esteroides/fisiología , Factores de Transcripción/fisiología , Animales , Células Cultivadas , Humanos , Ratones , Receptor X de Pregnano
2.
J Hepatol ; 51(1): 114-26, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19443070

RESUMEN

BACKGROUND/AIMS: The aim of this study was to identify human liver proteins that are associated with different stages of liver development. METHODS: We collected liver samples from 14 fetuses between 14 and 41 weeks of development, one child and four adults. Proteins which exhibited consistent and significant variations during development by two-dimensional differential in gel electrophoresis (2D-DIGE) were subjected to peptide mass fingerprint analysis by MALDI-TOF mass spectrometry. Real-time PCR analysis confirmed, at the transcriptional level, the data obtained by the proteomic approach. RESULTS: Among a total of 80 protein spots showing differential expression, we identified 42 different proteins or polypeptide chains, of which 26 were upregulated and 16 downregulated in developing in comparison to adult liver. These proteins could be classified in specific groups according to their function. By comparing their temporal expression profiles, we identified protein groups that were associated with different developmental stages of human fetal liver and suggest that the changes in protein expression observed during the 20- to 36-week time window play a pivotal role in liver development. CONCLUSIONS: The identification of these proteins may represent good markers of human liver and stem cells differentiation.


Asunto(s)
Electroforesis en Gel Bidimensional/métodos , Hígado/química , Hígado/embriología , Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Adulto , Canales de Calcio/análisis , Canales de Calcio/fisiología , Chaperonina con TCP-1 , Chaperoninas/análisis , Chaperoninas/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular , Hígado/metabolismo , Proteínas/análisis , Proteínas/fisiología , ARN Mensajero/análisis , Canales Catiónicos TRPV/análisis , Canales Catiónicos TRPV/fisiología
3.
J Clin Invest ; 115(1): 177-86, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15630458

RESUMEN

Vitamin D controls calcium homeostasis and the development and maintenance of bones through vitamin D receptor activation. Prolonged therapy with rifampicin or phenobarbital has been shown to cause vitamin D deficiency or osteomalacia, particularly in patients with marginal vitamin D stores. However, the molecular mechanism of this process is unknown. Here we show that these drugs lead to the upregulation of 25-hydroxyvitamin D(3)-24-hydroxylase (CYP24) gene expression through the activation of the nuclear receptor pregnane X receptor (PXR; NR1I2). CYP24 is a mitochondrial enzyme responsible for inactivating vitamin D metabolites. CYP24 mRNA is upregulated in vivo in mice by pregnenolone 16alpha-carbonitrile and dexamethasone, 2 murine PXR agonists, and in vitro in human hepatocytes by rifampicin and hyperforin, 2 human PXR agonists. Moreover, rifampicin increased 24-hydroxylase activity in these cells, while, in vivo in mice, pregnenolone 16alpha-carbonitrile increased the plasma concentration of 24,25-dihydroxyvitamin D(3). Transfection of PXR in human embryonic kidney cells resulted in rifampicin-mediated induction of CYP24 mRNA. Analysis of the human CYP24 promoter showed that PXR transactivates the sequence between -326 and -142. We demonstrated that PXR binds to and transactivates the 2 proximal vitamin D-responsive elements of the human CYP24 promoter. These data suggest that xenobiotics and drugs can modulate CYP24 gene expression and alter vitamin D(3) hormonal activity and calcium homeostasis through the activation of PXR.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Regulación de la Expresión Génica , Osteomalacia/inducido químicamente , Osteomalacia/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Esteroide Hidroxilasas/genética , Animales , Células Cultivadas , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/sangre , Sistema Enzimático del Citocromo P-450/química , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Mutación/genética , Osteomalacia/metabolismo , Receptor X de Pregnano , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores de Esteroides/agonistas , Rifampin/farmacología , Esteroide Hidroxilasas/biosíntesis , Esteroide Hidroxilasas/sangre , Esteroide Hidroxilasas/química , Activación Transcripcional/genética , Elemento de Respuesta a la Vitamina D/genética , Vitamina D3 24-Hidroxilasa
4.
Med Sci (Paris) ; 24(3): 301-5, 2008 Mar.
Artículo en Francés | MEDLINE | ID: mdl-18334180

RESUMEN

Decreased drug metabolism, hyperbilirubinemia and intrahepatic cholestasis are frequently observed during inflammation. Additionally, it has long been appreciated that exposure to drug metabolism-inducing xenobiotics can impair immune function. The nuclear receptor CAR (constitutive androstane receptor or NR1I3) and PXR (pregnane X receptor, NR1I2) control phase I (cytochrome P450 2B and 3A), phase II (GSTA, UGT1A1), and transporter (MDR1, SLC21A6, MRP2) genes involved in drugs metabolism, bile acids and bilirubin clearance in response to xenobiotics. It is well known that inflammation, through the activation of NF-kappaB pathway, leads to a decrease of CAR, PXR and RXRalpha expression and the expression of their target genes. In addition, a new study reveals the mutual repression between PXR and NF-kappaB signaling pathways, providing a molecular mechanism linking xenobiotic metabolism and inflammation.


Asunto(s)
Biotransformación/fisiología , Inflamación/metabolismo , FN-kappa B/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Esteroides/fisiología , Factores de Transcripción/fisiología , Xenobióticos/farmacocinética , Animales , Ácidos y Sales Biliares/metabolismo , Bilirrubina/metabolismo , Receptor de Androstano Constitutivo , Citocinas/fisiología , Regulación de la Expresión Génica/fisiología , Glucocorticoides/fisiología , Humanos , Hiperbilirrubinemia/etiología , Inactivación Metabólica/fisiología , Infecciones/inmunología , Infecciones/metabolismo , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/fisiopatología , Hígado/metabolismo , Tasa de Depuración Metabólica , Ratones , Ratones Noqueados , Receptor X de Pregnano , Transducción de Señal/fisiología
5.
Free Radic Biol Med ; 43(10): 1439-52, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17936189

RESUMEN

Vitamin E (alpha-tocopherol) has demonstrated antioxidant activity and gene-regulatory properties. d-Galactosamine (D-GalN)-induced cell death is mediated by nitric oxide in hepatocytes, and it is associated with hepatic steatosis. The beneficial properties of alpha-tocopherol and their relation to oxidative stress and gene regulation were assessed in D-GalN-induced cell death. Hepatocytes were isolated from human liver resections by a collagenase perfusion technique. alpha-Tocopherol (50 microM) was administered at the advanced stages (10 h) of D-GalN-induced cell death in cultured hepatocytes. Cell death, oxidative stress, alpha-tocopherol metabolism, and NF-kappaB-, pregnane X receptor (PXR)-, and peroxisome proliferator-activated receptor (PPAR-alpha)-associated gene regulation were estimated in the hepatocytes. D-GalN increased cell death and alpha-tocopherol metabolism. alpha-Tocopherol exerted a moderate beneficial effect against apoptosis and necrosis induced by D-GalN. Induction (rifampicin) or inhibition (ketoconazole) of alpha-tocopherol metabolism and overexpression of PXR showed that the increase in PXR-related CYP3A4 expression caused by alpha-tocopherol enhanced cell death in hepatocytes. Nevertheless, the reduction in NF-kappaB activation and inducible nitric oxide synthase expression and the enhancement of PPAR-alpha and carnitine palmitoyl transferase gene expression by alpha-tocopherol may be relevant for cell survival. In conclusion, the cytoprotective properties of alpha-tocopherol are mostly related to gene regulation rather than to antioxidant activity in toxin-induced cell death in hepatocytes.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Citoprotección , Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , alfa-Tocoferol/farmacología , Apoptosis/genética , Carnitina O-Palmitoiltransferasa/genética , Células Cultivadas , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/metabolismo , Galactosamina/antagonistas & inhibidores , Galactosamina/toxicidad , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Humanos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , PPAR alfa/metabolismo , Receptor X de Pregnano , Especies Reactivas de Oxígeno/metabolismo , Receptores de Esteroides/metabolismo , alfa-Tocoferol/metabolismo
6.
Eur J Cancer ; 42(6): 785-92, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16510279

RESUMEN

In the last few years, several studies have provided a causal link between constitutive activation of nuclear factor kappa-B (NF-kappaB) and the initiation and development of cancer. More recently, it appears that a cancer-induced inflammatory response may be an important factor in the inter-individual variability of the response to and toxic effects of cancer chemotherapy, as well as in the alteration of drug metabolism enzyme expression in patients. The relationships between chronic inflammation (or infection), cancer and drug metabolism are many: chronic infections lead to inflammation, inflammation may lead to cancer, cancer usually leads to an inflammatory syndrome, and inflammation alters the expression of drug metabolising enzymes and thus of the efficiency of cancer chemotherapy. This review focuses on the functional consequences of NF-kappaB activation during oncogenesis and on the expression of the major cytochrome P450s (CYP) involved in anticancer therapies. Finally, the potential role of NF-kappaB as the missing link between inflammation, cancer and alteration in hepatic drug metabolism in patients with cancer is discussed.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Inflamación/metabolismo , Hígado/metabolismo , FN-kappa B/fisiología , Neoplasias/metabolismo , Expresión Génica , Humanos , Inflamación/complicaciones , Neoplasias/patología
7.
Cell Signal ; 17(2): 187-96, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15494210

RESUMEN

The role of microtubules (MTs) in steroid hormone-dependent human glucocorticoid receptor (hGR) activation/translocation is controversial. It was demonstrated recently that colchicine (COL) down-regulates hGR-driven genes in primary human hepatocytes by a mechanism involving inhibition of hGR translocation to the nucleus. To investigate whether inhibition of hGR translocation is the sole reason for its inactivation, we used human cervical carcinoma cells (HeLa) as a model. Herein we present evidence that perturbation of microtubules in HeLa cells leads to rapid time- and dose-dependent degradation of hGR protein. Degradation is proteasome mediated as revealed by its reversibility by proteasome inhibitor MG132. Moreover, degradation was observed for neither wt-hGR nor hGR mutants S226A and K419A in transiently transfected COS-1 cells. On the other hand, c-jun N-terminal kinase (JNK) seems not to be involved in the process because JNK inhibitor 1,9-Pyrazoloanthrone (SP600125) does not reverse hGR degradation. Similarly, another hGR functional antagonist, nuclear factor kappa beta (NFkappaB), did not play any role in the degradation process.


Asunto(s)
Microtúbulos/fisiología , Receptores de Glucocorticoides/metabolismo , Animales , Antracenos/farmacología , Células COS , Chlorocebus aethiops , Colchicina/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Citosol/metabolismo , Dexametasona/farmacología , Inhibidores Enzimáticos/farmacología , Células HeLa , Humanos , Espacio Intranuclear/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leupeptinas/farmacología , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mutación/fisiología , FN-kappa B/metabolismo , Nocodazol/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptores de Glucocorticoides/genética , Transfección , Ubiquitina/metabolismo , Vincristina/farmacología
8.
Mol Endocrinol ; 17(1): 42-55, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12511605

RESUMEN

The constitutive androstane receptor (CAR, NR1I3) transcriptionally activates cytochrome P450 2B6, 2C9, and 3A4 when activated by xenobiotics, such as phenobarbital. Information on the human CAR promoter was obtained by searching the NCBI human genome database. A contig (NT026945) corresponding to a fragment of chromosome 1q21 was found to contain the complete CAR gene. These data were confirmed using chromosomal in situ hybridization. Both primer extension and 5'-rapid amplification of the cDNA end PCR analysis were carried out to determine the transcriptional start site of human CAR, which was found to be 32 nucleotides downstream of a potential TATA box (CATAAAA). In addition, we found that the 5'-untranslated region of CAR mRNA is 110 nucleotides shorter than previously reported. Using genomic PCR, we amplified and cloned approximately 4.9 kb (-4711/+144) of the CAR gene promoter. The activity of this promoter was measured by transient transfection. Deletion analysis suggested the presence of a glucocorticoid responsive element in its distal region (-4477/-4410). From cotransfection experiments, mutagenesis, and gel shift assays, we identified a glucocorticoid response element at -4447/-4432 that was recognized and transactivated by the human glucocorticoid receptor. Finally, using the chromatin immunoprecipitation assay, we demonstrated that the glucocorticoid receptor binds to the distal region of CAR promoter in cultured hepatocytes only in the presence of dexamethasone. Identification of this functional element provides a rational mechanistic basis for CAR induction by glucocorticoids. CAR appears to be a primary glucocorticoid receptor-response gene.


Asunto(s)
Regulación de la Expresión Génica , Regiones Promotoras Genéticas/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Glucocorticoides/genética , Elementos de Respuesta/genética , Factores de Transcripción/genética , Células Cultivadas , Clonación Molecular , Receptor de Androstano Constitutivo , Hepatocitos , Humanos , ARN Mensajero/análisis , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/aislamiento & purificación , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/aislamiento & purificación , Factores de Transcripción/química , Factores de Transcripción/aislamiento & purificación , Transcripción Genética , Activación Transcripcional/fisiología
9.
Mol Endocrinol ; 17(9): 1693-703, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12805410

RESUMEN

SHP (small heterodimer partner, NR1I0) is an atypical orphan member of the nuclear receptor subfamily in that it lacks a DNA-binding domain. It is mostly expressed in the liver, where it binds to and inhibits the function of nuclear receptors. SHP is up-regulated by primary bile acids, through the activation of their receptor farnesoid X receptor, leading to the repression of cholesterol 7alpha-hydroxylase (CYP7alpha) expression, the rate-limiting enzyme in bile acid production from cholesterol. PXR (pregnane X receptor, NR1I2) is a broad-specificity sensor that recognizes a wide variety of synthetic drugs as well as endogenous compounds such as bile acid precursors. Upon activation, PXR induces CYP3A and inhibits CYP7alpha, suggesting that PXR can act on both bile acid synthesis and elimination. Indeed, CYP7alpha and CYP3A are involved in biochemical pathways leading to cholesterol conversion into primary bile acids, whereas CYP3A is also involved in the detoxification of toxic secondary bile acid derivatives. Here, we show that PXR is a target for SHP. Using pull-down assays, we show that SHP interacts with both murine and human PXR in a ligand-dependent manner. From transient transfection assays, SHP is shown to be a potent repressor of PXR transactivation. Furthermore, we report that chenodeoxycholic acid and cholic acid, two farnesoid X receptor ligands, induce up-regulation of SHP and provoke a repression of PXR-mediated CYP3A induction in human hepatocytes as well as in vivo in mice. These results reveal an elaborate regulatory cascade, tightly controlled by SHP, for both the maintenance of bile acid production and detoxification in the liver.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Hepatocitos/metabolismo , Histona Acetiltransferasas , Humanos , Ligandos , Ratones , Coactivador 1 de Receptor Nuclear , Receptor X de Pregnano , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología
12.
Endocrinology ; 151(4): 1653-61, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20185760

RESUMEN

The pregnane X receptors (PXRs) and the constitutive androstane receptor (CAR) were initially isolated as nuclear receptors regulating xenobiotic metabolism and elimination, alleviating chemical insults. However, recent works suggest that these xenoreceptors play an endobiotic role in modulating hepatic lipid metabolism. In this study, we show that CAR activators]phenobarbital and 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime] induce the lipogenic gene thyroid hormone-responsive spot 14 protein (THRSP) (or Spot14, S14) expression in human hepatocytes. In addition, we report that treatment of wild-type mice with mCAR activators (phenobarbital and 1,4-Bis[2-(3,5-dichloropyridyloxy)]benzene) efficiently increases thrsp expression, in contrast to CAR null mice. We demonstrate that CAR directly transactivates THRSP promoter through the direct repeat with 4-bp spacer thyroid hormone and PXR response element. Deletion or point mutations within this PXR response element led to a drastic inhibition of CAR-mediated THRSP transactivation. Gel-shift analysis revealed that the CAR/retinoid X receptor complex binds to this element. In conclusion, our results indicate that THRSP gene is a CAR and PXR target gene. Because THRSP expression correlates with lipogenesis and insulin sensitivity, our data suggest that CAR and/or PXR activating drugs and xenobiotics may promote aberrant hepatic de novo lipogenesis leading potentially to fatty liver diseases and insulin resistance.


Asunto(s)
Hígado/metabolismo , Proteínas Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Adulto , Anciano , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Receptor de Androstano Constitutivo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Masculino , Ratones , Persona de Mediana Edad , Proteínas Nucleares/genética , Oximas/farmacología , Fenobarbital/farmacología , Regiones Promotoras Genéticas/genética , Piridinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiazoles/farmacología , Factores de Transcripción/genética , Activación Transcripcional , Transfección
13.
Mol Pharm ; 5(1): 35-41, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18159929

RESUMEN

Xenobiotic and drug metabolism and transport are managed by a large number of genes coordinately regulated by at least three nuclear receptors or xenosensors: aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR, NR1I3), and pregnane X receptor (PXR, NR1I2). Initially characterized as xenosensors, it is now evident that CAR and PXR also trigger pleiotropic effects on liver function. Recent studies have shown the existence of crosstalk between xenosensors and other nuclear receptors or transcription factors controlling endogenous signaling pathways which regulate physiological functions. This review is focused on recent observations showing that activation of CAR and PXR alters lipid metabolism, glucose homeostasis, and inflammation by interfering with HNF4alpha, FoxO1, FoxA2, PGC1alpha, or NFkB p65. Such crosstalks explain clinical observations and provide molecular mechanisms allowing understanding how xenobiotics and drugs may affect physiological functions and provoke endocrine disruptions.


Asunto(s)
Glucosa/metabolismo , Metabolismo de los Lípidos , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Factores de Transcripción/metabolismo , Animales , Receptor de Androstano Constitutivo , Humanos , Inflamación , Receptor X de Pregnano , Síndrome de Respuesta Inflamatoria Sistémica/etiología
14.
Artículo en Inglés | MEDLINE | ID: mdl-17608617

RESUMEN

The expression of many genes involved in xenobiotic/drug metabolism and transport is regulated by at least three nuclear receptors or xenosensors: aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR), and pregnane X receptor (PXR). These receptors establish crosstalk with other nuclear receptors or transcription factors controlling signaling pathways that regulate the homeostasis of bile acids, lipids, glucose, inflammation, vitamins, hormones, and others. These crosstalks are expected to modify profoundly our vision of xenobiotic/drug disposition and toxicity. They provide molecular mechanisms to explain how physiopathological stimuli affect xenobiotic/drug disposition, and how xenobiotics/drugs may affect physiological functions and generate toxic responses. In addition, the possibility that xenosensors may control other signaling pathways opens the way to new pharmacological opportunities.


Asunto(s)
Regulación de la Expresión Génica , Receptores Citoplasmáticos y Nucleares/metabolismo , Xenobióticos/farmacocinética , Transporte Biológico/fisiología , Homeostasis/fisiología , Humanos , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Xenobióticos/efectos adversos
15.
Biochem Biophys Res Commun ; 360(1): 76-82, 2007 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-17585873

RESUMEN

We previously reported that the pregnane X receptor (PXR) interferes with vitamin D receptor (VDR) target genes, notably CYP24, by targeting the same responsive elements. Since PXR and constitutive androstane receptor (CAR) share responsive elements in the promoter of their target genes, we wondered whether CAR also interferes with CYP24 expression. The current study shows that: (i) CAR-RXR heterodimer binds to and transactivates the proximal promoter of CYP24 (-1200/+22) and both VDRE-1 and VDRE-2 which control its expression in response to 1,25-dihydroxyvitamin D(3), (ii) androstanol an inverse agonist of hCAR inhibits transactivation of VDREs by hCAR, (iii) mutations of either VDRE-1 or -2 half sites inhibit hCAR-mediated transactivation, and (iv) in primary human hepatocytes (n =11) CITCO, a specific hCAR agonist, is an inducer of CYP24 as well as of CYP2B6 and CYP3A4 mRNAs. In conclusion, CAR/PXR and VDR bind to and transactivate the same response elements in CYP24 promoter.


Asunto(s)
Expresión Génica/fisiología , Hepatocitos/metabolismo , Receptores de Calcitriol/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología , Esteroide Hidroxilasas/metabolismo , Factores de Transcripción/metabolismo , Células Cultivadas , Receptor de Androstano Constitutivo , Humanos , Vitamina D3 24-Hidroxilasa
16.
Hepatology ; 45(5): 1146-53, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17464991

RESUMEN

Constitutive androstane receptor (CAR; NR1I3) controls the metabolism and elimination of endogenous and exogenous toxic compounds by up-regulating a battery of genes. In this work, we analyzed the expression of human CAR (hCAR) in normal liver during development and in hepatocellular carcinoma (HCC) and investigated the effect of hepatocyte nuclear factor 4alpha isoforms (HNF4alpha1 and HNF4alpha7) on the hCAR gene promoter. By performing functional analysis of hCAR 5'-deletions including mutants, chromatin immunoprecipitation in human hepatocytes, electromobility shift and cotransfection assays, we identified a functional and species-conserved HNF4alpha response element (DR1: ccAGGCCTtTGCCCTga) at nucleotide -144. Both HNF4alpha isoforms bind to this element with similar affinity. However, HNF4alpha1 strongly enhanced hCAR promoter activity whereas HNF4alpha7 was a poor activator and acted as a repressor of HNF4alpha1-mediated transactivation of the hCAR promoter. PGC1alpha stimulated both HNF4alpha1-mediated and HNF4alpha7-mediated hCAR transactivation to the same extent, whereas SRC1 exhibited a marked specificity for HNF4alpha1. Transduction of human hepatocytes by HNF4alpha7-expressing lentivirus confirmed this finding. In addition, we observed a positive correlation between CAR and HNF4alpha1 mRNA levels in human liver samples during development, and an inverse correlation between CAR and HNF4alpha7 mRNA levels in HCC. These observations suggest that HNF4alpha1 positively regulates hCAR expression in normal developing and adult livers, whereas HNF4alpha7 represses hCAR gene expression in HCC.


Asunto(s)
Factor Nuclear 4 del Hepatocito/fisiología , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Factores de Transcripción/biosíntesis , Carcinoma Hepatocelular/metabolismo , Línea Celular , Receptor de Androstano Constitutivo , Regulación de la Expresión Génica , Humanos , Hígado/crecimiento & desarrollo , Neoplasias Hepáticas/metabolismo , Regiones Promotoras Genéticas/fisiología , Isoformas de Proteínas/fisiología , Elementos de Respuesta/fisiología
17.
Mol Pharmacol ; 70(1): 329-39, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16608920

RESUMEN

The constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) play a major part in the control of drug metabolism and transport. We have previously shown that PXR and CAR expression is controlled by the glucocorticoid receptor (GR) and proposed the existence of a signal transmission cascade GR-(PXR/CAR)-drug metabolizing and transporter systems. In the current study, we investigated the effect of ketoconazole and other azole-derived drugs, miconazole and fluconazole, on the transcriptional activity of the human GR (hGR) in HeLa and HepG2 cells, and in primary human hepatocytes. The data show that ketoconazole inhibits GR transcriptional activity and competes with dexamethasone for hGR binding. In primary human hepatocytes, ketoconazole inhibits the expression of 1) GR-responsive genes tyrosine aminotransferase and both PXR and CAR; 2) CAR and PXR target genes, including cytochromes P450 (P450) CYP2B6, CYP2C9, and CYP3A4; UDP-glucuronosyltransferase 1A1, glutathione S-transferases A1 and A2; and transporter proteins (phase III) solute carrier family 21 form A6 and multidrug resistance protein 2. In parallel experiments, ketoconazole affected neither the expression of GR, the expression of glyceraldehyde-3-phosphate dehydrogenase, nor the inducible expression of CYP1A1 and 1A2. Miconazole behaved like ketoconazole, whereas fluconazole had no effect. We conclude that, in addition to their well known inhibitory effect on P450 enzyme activities, ketoconazole and miconazole are antagonists of hGR. These results provide a novel molecular mechanism by which these compounds may exert adverse and toxic effects on drug metabolism and other functions in human.


Asunto(s)
Cetoconazol/farmacología , Miconazol/farmacología , Receptores de Glucocorticoides/antagonistas & inhibidores , Adulto , Anciano , Northern Blotting , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Receptor de Androstano Constitutivo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Células HeLa , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Persona de Mediana Edad , Receptor X de Pregnano , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Eur J Biochem ; 270(3): 556-64, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12542705

RESUMEN

Induction of CYP3A genes by the ligand-activated pregnane-X-receptor (PXR) involves the interaction of other as yet unidentified liver transcription factors. Here we show that the CYP3A1 promoter contains two active sites controlled by the CCAAT/enhancer-binding protein alpha (C/EBPalpha), previously shown to regulate a number of liver stress response genes. We have identified two functional C/EBP binding sites at the CYP3A1 promoter that confer luciferase activity to C/EBPalpha cotransfected CHO cells. When inserted upstream of a thymidine kinase promoter, oligonucleotides corresponding to these elements (-350/-311 and -628/-608), increase reporter gene expression when cotransfected with a C/EBPalpha expression vector. Point mutations in the most conserved nucleotides in either element prevent binding of C/EBPalpha and abolish transactivation of the CYP3A1 promoter. Moreover, we demonstrate that C/EBPalpha accumulates in the rat liver nuclei in response to dexamethasone, and that under these conditions C/EBPalpha binds to the CYP3A1 promoter elements. Our results suggest a correlation between transcription of C/EBPalpha, nuclear protein function and induction of CYP3A1 by dexamethasone in the liver. They also support the notion that C/EBPalpha participates in the up-regulation of the CYP3A1 gene in response to synthetic glucocorticoids.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Secuencia de Bases , Sitios de Unión , Western Blotting , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Citocromo P-450 CYP3A , Ensayo de Cambio de Movilidad Electroforética , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacología , Humanos , Masculino , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Plásmidos , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/fisiología , Elementos de Respuesta , Transfección
19.
J Biol Chem ; 277(28): 25125-32, 2002 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-11991950

RESUMEN

The fully active dihydroxylated metabolite of vitamin D(3) induces the expression of CYP3A4 and, to a lesser extent, CYP2B6 and CYP2C9 genes in normal differentiated primary human hepatocytes. Electrophoretic mobility shift assays and cotransfection in HepG2 cells using wild-type and mutated oligonucleotides revealed that the vitamin D receptor (VDR) binds and transactivates those xenobiotic-responsive elements (ER6, DR3, and DR4) previously identified in CYP3A4, CYP2B6, and CYP2C9 promoters and shown to be targeted by the pregnane X receptor (PXR) and/or the constitutive androstane receptor (CAR). Full VDR response of various CYP3A4 heterologous/homologous promoter-reporter constructs requires both the proximal ER6 and the distal DR3 motifs, as observed previously with rifampicin-activated PXR. Cotransfection of a CYP3A4 homologous promoter-reporter construct (including distal and proximal PXR-binding motifs) and of PXR or CAR expression vectors in HepG2 cells revealed the ability of these receptors to compete with VDR for transcriptional regulation of CYP3A4. In conclusion, this work suggests that VDR, PXR, and CAR control the basal and inducible expression of several CYP genes through competitive interaction with the same battery of responsive elements.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas , Sistema Enzimático del Citocromo P-450/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Hepatocitos/metabolismo , Oxigenasas de Función Mixta/genética , Oxidorreductasas N-Desmetilantes/genética , Receptores de Calcitriol/fisiología , Esteroide 16-alfa-Hidroxilasa , Esteroide Hidroxilasas/genética , Secuencia de Bases , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A , Cartilla de ADN , Hepatocitos/enzimología , Humanos , Receptores de Calcitriol/metabolismo , Células Tumorales Cultivadas
20.
J Biol Chem ; 277(1): 209-17, 2002 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-11679585

RESUMEN

Although cytochrome P450 2C9 (CYP2C9) is a major CYP expressed in the adult human liver, its mechanism of regulation is poorly known. In previous work, we have shown that CYP2C9 is inducible in primary human hepatocytes by xenobiotics including dexamethasone, rifampicin, and phenobarbital. The aim of this work was to investigate the molecular mechanism(s) controlling the inducible expression of CYP2C9. Deletional analysis of CYP2C9 regulatory region (+21 to -2088) in the presence of various hormone nuclear receptors suggested the presence of two functional response elements, a glucocorticoid receptor-responsive element (-1648/-1684) and a constitutive androstane receptor-responsive element (CAR, -1783/-1856). Each of these were characterized by co-transfection experiments, directed mutagenesis, gel shift assays, and response to specific antagonists RU486 and androstanol. By these experiments we located a glucocorticoid-responsive element imperfect palindrome at -1662/-1676, and a DR4 motif at -1803/-1818 recognized and transactivated by human glucocorticoid receptor and by hCAR and pregnane X receptor, respectively. Identification of these functional elements provides rational mechanistic basis for CYP2C9 induction by dexamethasone (submicromolar concentrations), and by phenobarbital and rifampicin, respectively. CYP2C9 appears therefore to be a primary glucocorticoid-responsive gene, which in addition, may be induced by xenobiotics through CAR/pregnane X receptor activation.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas , Sistema Enzimático del Citocromo P-450/genética , Regulación Enzimológica de la Expresión Génica , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Glucocorticoides/fisiología , Esteroide 16-alfa-Hidroxilasa , Esteroide Hidroxilasas/genética , Factores de Transcripción/fisiología , Células Cultivadas , Receptor de Androstano Constitutivo , Cicloheximida/farmacología , Citocromo P-450 CYP2C9 , Hepatocitos/enzimología , Humanos , Receptor X de Pregnano , ARN Mensajero/análisis , Receptores de Ácido Retinoico/fisiología , Receptores de Esteroides/fisiología , Elementos de Respuesta , Receptores X Retinoide , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA