Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Endocrinol ; 188(1): 91-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16394178

RESUMEN

The induction of vascular endothelial growth factor (VEGF) expression by 17beta-estradiol (E(2)) in many target cells, including epithelial cells, fibroblasts and smooth muscle cells, suggests a role for this hormone in the modulation of angiogenesis and vascular permeability. We have already described a cyclic increase in Flk-1/KDR-expressing capillaries in the human endometrium during the proliferative and mid-secretory phases, strongly suggestive of an E(2) effect on Flk-1/KDR expression in the endometrial capillaries. However, it is unclear whether these processes are due to a direct effect of E(2) on endothelial cells. Using immunohistochemistry, we report an increase in Flk-1/KDR expression in endometrial capillaries of ovariectomized mice treated with E(2), or both E(2) and progesterone. This process is mediated through estrogen receptor (ER) activation. In vitro experiments using quantitative RT-PCR analysis demonstrate that Flk-1/KDR expression was not regulated by E(2) in human endothelial cells from the microcirculation (HMEC-1) or macrocirculation (HUVEC), even in endothelial cells overexpressing ERalpha or ERbeta after ER-mediated adenovirus infection. In contrast, Flk-1/KDR expression was up-regulated by VEGF itself, in a time- and dose-dependent manner, with the maximal response at 10 ng/ml. Thus, we suggest that E(2) up-regulates Flk-1/KDR expression in vivo in endothelial cells mainly through the modulation of VEGF by a paracrine mechanism. It is currently unknown whether or not the endothelial origin might account for differences in the E(2)-modulation of VEGF receptor expression, particularly in relation to the vascular bed of sex steroid-responsive tissues.


Asunto(s)
Estradiol/farmacología , Transducción de Señal/efectos de los fármacos , Útero/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/análisis , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/análisis , Femenino , Humanos , Inmunohistoquímica/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Neovascularización Fisiológica , Ovariectomía , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba , Útero/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
2.
Oncogene ; 25(12): 1799-806, 2006 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-16158045

RESUMEN

The proliferative action of ERalpha largely accounts for the carcinogenic activity of estrogens. By contrast, recent data show that ERbeta displays tumor-suppressor properties, thus supporting the interest to identify compounds that could increase its activity. Here, we show that histone deacetylase inhibitors (HDI) upregulated ERbeta protein levels, whereas it decreased ERalpha expression. Part of this regulation took place at the mRNA level through a mechanism independent of de novo protein synthesis. In addition, we found that, in various cancer cells, the treatment with different HDI enhanced the ligand-dependent activity of ERbeta more strongly than that of ERalpha. On the other hand, in MDA-MB231 and HeLa cells, the expression of ERs modified the transcriptional response to HDI. The use of deletion mutants of both receptors demonstrated that AF1 domain of the receptors was required. Finally, we show that ERbeta expression led to a dramatic increased in the antiproliferative activity of HDI, which correlated with a modification of the transcription of genes involved in cell cycle control by HDI. Altogether, these data demonstrate that the interference of ERbeta and HDAC on the control of transcription and cell proliferation constitute a promising approach for cancer therapy.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/efectos de los fármacos , Histona Desacetilasas/metabolismo , Transcripción Genética/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Proliferación Celular/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Células HeLa , Histona Desacetilasas/efectos de los fármacos , Humanos , Reacción en Cadena de la Polimerasa , ARN Mensajero
3.
Endocr Relat Cancer ; 11(3): 537-51, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15369453

RESUMEN

The characterization of estrogen receptor beta (ERbeta) brought new insight into the mechanisms underlying estrogen signaling. Estrogen induction of cell proliferation is a crucial step in carcinogenesis of gynecologic target tissues, and the mitogenic effects of estrogen in these tissues (such as breast, endometrium and ovary) are well documented both in vitro and in vivo. There is also an emerging body of evidence that colon and prostate cancer growth is influenced by estrogens. In all of these tissues, most studies have shown decreased ERbeta expression in cancer as compared with benign tumors or normal tissues, whereas ERalpha expression persists. The loss of ERbeta expression in cancer cells could reflect tumor cell dedifferentiation but may also represent a critical stage in estrogen-dependent tumor progression. Modulation of the expression of ERalpha target genes by ERbeta or ERbeta-specific gene induction could explain that ERbeta has a differential effect on proliferation as compared with ERalpha. ERbeta may exert a protective effect and thus constitute a new target for hormone therapy, such as ligand specific activation. The potential distinct roles of ERalpha and ERbeta expression in carcinogenesis, as suggested by experimental and clinical data, are discussed in this review.


Asunto(s)
Neoplasias de la Mama/etiología , Receptor beta de Estrógeno/deficiencia , Neoplasias Hormono-Dependientes/etiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Receptor alfa de Estrógeno/deficiencia , Femenino , Humanos , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología
4.
J Endocrinol ; 179(1): 41-53, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14529564

RESUMEN

We analysed the antiproliferative activity of various histone deacetylase (HDAC) inhibitors such as trichostatin A (TSA) on human breast cancer cells. We observed a lower sensitivity to HDAC inhibition for oestrogen receptor negative (ER-) versus positive (ER+) cell lines. This differential response was associated neither with a modification of drug efflux via the multidrug resistance system nor with a global modification of histone acetyltransferase (HAT)/HDAC activities. In contrast, we demonstrated that in ER+ breast cancer cells the p21(WAF1/CIP1) gene was more sensitive to TSA regulation and was expressed at higher levels. These differences were observed both in transient transfection experiments and on the endogenous p21(WAF1/CIP1) gene. The Sp1 transcription factor, which was shown to interact in vitro with both class I and class II HDACs, is sufficient to confer the differential sensitivity to TSA and participated in the control of p21(WAF1/CIP1) basal expression. Finally, re-expression of ERalpha following adenoviral infection of ER- breast cancer cells increased both p21(WAF1/CIP1) protein accumulation and the growth inhibitory activity of TSA. Altogether, our results highlight the key role of ERalpha and p21(WAF1/CIP1) gene expression in the sensitivity of breast cancer cells to hyperacetylating agents.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Ciclinas/genética , Inhibidores de Histona Desacetilasas , Receptores de Estrógenos/fisiología , Acetilación/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Receptor alfa de Estrógeno , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores de Estrógenos/metabolismo , Factor de Transcripción Sp1/fisiología , Transfección , Células Tumorales Cultivadas
5.
Endocrinology ; 142(9): 4120-30, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11517191

RESUMEN

Recent studies indicate that the expression of ER beta in breast cancer is lower than in the normal breast, suggesting that ER beta could play an important role in carcinogenesis. To investigate this hypothesis, we engineered ER-negative MDA-MB-231 (human breast cancer cells) to reintroduce either ER alpha or ER beta protein with an adenoviral vector. In these cells, ER beta (as ER alpha) expression was monitored using RT-PCR and Western blot. ER beta protein was localized in the nucleus (immunocytochemistry) and able to transactivate estrogen-responsive reporter constructs in the presence of E2. ER beta and ER alpha induced the expression of several endogenous genes such as pS2, TGF alpha, or the cyclin kinase inhibitor p21 but, in contrast to ER alpha, ER beta was unable to regulate c-myc proto-oncogene expression. The pure antiestrogen ICI 164, 384 completely blocked ER alpha and ER beta estrogen-induced activities. ER beta inhibited MDA-MB-231 cell proliferation in a ligand-independent manner, whereas ER alpha inhibition of proliferation is hormone dependent. Moreover, ER beta and ER alpha decreased cell motility and invasion. Our data bring the first evidence that ER beta is an important modulator of proliferation and invasion of breast cancer cells and support the hypothesis that the loss of ER beta expression could be one of the events leading to the development of breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Receptores de Estrógenos/fisiología , Adenoviridae/genética , División Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Estrógenos/fisiología , Regulación de la Expresión Génica/fisiología , Técnicas de Transferencia de Gen , Genes Reporteros/fisiología , Vectores Genéticos , Humanos , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Proto-Oncogenes Mas
6.
J Steroid Biochem Mol Biol ; 77(4-5): 193-203, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11457657

RESUMEN

Estrogen receptor (ER) and cAMP signaling pathways interact in a number of estrogen target tissues including mammary and uterine tissues. One aspect of this interaction is that estradiol and protein kinase A (PKA) activators can cooperate synergistically to activate ER-mediated transcription of both endogenous genes and reporter genes containing only estrogen response elements. The purpose of this study was to investigate the molecular mechanism of this interaction between signaling pathways. Site-directed mutagenesis of the potential PKA phosphorylation sites in the ER indicated that phosphorylation of these sites was not necessary for the observed transcriptional synergy. In transient transfection assays in two different cell lines using reporter constructs containing either cAMP response elements, estrogen response elements or both types of elements, with the addition or absence of cAMP response element binding protein (CREB) expression plasmid, we observed that only one of these cell lines exhibited estrogen/PKA transcriptional synergy. Experiments demonstrated that CREB itself was involved in the transcriptional synergy, and that transfection of CREB restored transcriptional synergy in the cell line in which it was lacking. A functional interaction between ER and CREB was also demonstrated using a mammalian cell protein interaction assay; a dominant negative mutant of CREB did not exhibit this interaction. Therefore, these data indicate that CREB protein is required for the transcriptional synergy between cAMP and estrogen signaling pathways. Furthermore, CREB cooperated with the ER on genes that did not contain cAMP response elements, but contained only estrogen response elements. We propose that activated CREB is recruited to estrogen responsive genes by an ER--coactivator complex containing proteins such as CREB binding protein (CBP) and that the interaction of CREB with ER may assist in stabilizing its interaction with CBP and in promoting estrogen-ER and PKA transcriptional synergy.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN/metabolismo , Estradiol/farmacología , Receptores de Estrógenos/metabolismo , Animales , Células CHO , Cricetinae , ADN/efectos de los fármacos , Sinergismo Farmacológico , Activación Enzimática , Humanos , Mutagénesis Sitio-Dirigida , Fosforilación , Isoformas de Proteínas/metabolismo , Receptores de Estrógenos/genética , Transcripción Genética , Células Tumorales Cultivadas
7.
Mol Endocrinol ; 14(12): 1962-75, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11117527

RESUMEN

The nuclear peroxisome proliferator-activated receptors (PPARs) alpha, beta, and gamma activate the transcription of multiple genes involved in lipid metabolism. Several natural and synthetic ligands have been identified for each PPAR isotype but little is known about the phosphorylation state of these receptors. We show here that activators of protein kinase A (PKA) can enhance mouse PPAR activity in the absence and the presence of exogenous ligands in transient transfection experiments. Activation function 1 (AF-1) of PPARs was dispensable for transcriptional enhancement, whereas activation function 2 (AF-2) was required for this effect. We also show that several domains of PPAR can be phosphorylated by PKA in vitro. Moreover, gel retardation experiments suggest that PKA stabilizes binding of the liganded PPAR to DNA. PKA inhibitors decreased not only the kinase-dependent induction of PPARs but also their ligand-dependent induction, suggesting an interaction between both pathways that leads to maximal transcriptional induction by PPARs. Moreover, comparing PPAR alpha knockout (KO) with PPAR alpha WT mice, we show that the expression of the acyl CoA oxidase (ACO) gene can be regulated by PKA-activated PPAR alpha in liver. These data demonstrate that the PKA pathway is an important modulator of PPAR activity, and we propose a model associating this pathway in the control of fatty acid beta-oxidation under conditions of fasting, stress, and exercise.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Acil-CoA Oxidasa , Animales , Línea Celular , Toxina del Cólera/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ligandos , Masculino , Ratones , Ratones Noqueados , Modelos Genéticos , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Pirimidinas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Ácido Retinoico/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Receptores X Retinoide , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/agonistas , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Activación Transcripcional/efectos de los fármacos
8.
Recent Prog Horm Res ; 55: 163-93; discussion 194-5, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11036937

RESUMEN

The action of nuclear hormone receptors is tripartite, involving the receptor, its ligands, and its co-regulator proteins. The estrogen receptor (ER), a member of this superfamily, is a hormone-regulated transcription factor that mediates the effects of estrogens and anti-estrogens (e.g., tamoxifen) in breast cancer and other estrogen target cells. This chapter presents our recent work on several aspects of estrogen action and the function of the ER: 1) elucidation of ER structure-function relationships and development of ligands that are selective for one of the two ER subtypes, ERalpha or ERbeta; 2) identification of ER-selective co-regulators that potentiate the inhibitory effectiveness of anti-estrogens and dominant-negative ERs and modulate the activity of estrogens; 3) characterization of genes that are regulated by the anti-estrogen-ER versus the estrogen-ER complex; and 4) elucidation of the intriguing pharmacology of these ER complexes at different gene regulatory sites. These findings indicate that different residues of the ER hormone-binding domain are involved in the recognition of structurally distinct estrogens and anti-estrogens and highlight the exquisite precision of the regulation of ER activities by ligands, with small changes in ligand structure resulting in major changes in receptor character. Studies also explore the biology and distinct pharmacology mediated by ERalpha and ERbeta complexed with different ligands through different target genes. The upregulation of the anti-oxidant detoxifying phase II enzyme, quinone reductase, by the anti-estrogen-occupied ER, mediated via the electrophile response element in the QR gene, may contribute to the beneficial antioxidant effects of anti-estrogens in breast cancer and illustrates the activation of some genes by ER via non-estrogen response element sequences. The intriguing biology of estrogen in its diverse target cells is thus determined by the structure of the ligand, the ER subtype involved, the nature of the hormone-responsive gene promoter, and the character and balance of co-activators and co-repressors that modulate the cellular response to the ER-ligand complex. The continuing development of novel ligands and the study of how they function as selective agonists or antagonists through ERalpha or ERbeta should allow optimized tissue selectivity of these agents for hormone replacement therapy and treatment and prevention of breast cancer.


Asunto(s)
Receptores de Estrógenos/metabolismo , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Estrógenos/química , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Humanos , Ligandos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Prohibitinas , Receptores de Estrógenos/química , Receptores de Estrógenos/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
9.
Mol Endocrinol ; 13(6): 969-80, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10379895

RESUMEN

Dominant negative estrogen receptors are transcriptionally inactive, altered forms of the estrogen receptor (ER) that can dimerize with the ER and have the potential to inactivate the biological functions of this receptor. Here, we provide the first report that adenoviral delivery of a dominant negative ER to ER-positive breast cancer cells is able to effectively suppress estrogen-stimulated cell proliferation and the hormonal induction of endogenous genes. We constructed recombinant adenoviral vectors expressing a dominant negative ER (S554 fs, Ad-fs) or, for comparison, antisense ER (Ad-AS), or the sense wild-type ER (Ad-WT). Expression of the dominant negative ER or antisense ER, but not wild-type ER, blocked estradiol stimulation of the estrogen-responsive genes pS2 and c-myc. The dominant negative ER also fully abolished the estradiol-induced increase in proliferation of MCF-7 breast cancer cells, as did the antisense ER. The antiproliferative effects of the dominant negative and antisense ERs are explained by an increase in the number of cells in the G0/G1 stage of the cell cycle and decrease in the number of cells in G2/M as determined by flow cytometry, and also by a significant increase in the percentage of cells undergoing apoptosis. Our data strongly support the idea that targeting ER action using recombinant viral delivery of dominant negative ERs is an effective way to suppress ER-positive breast cancer cell proliferation and suggests the potential attractiveness of dominant negative gene therapy approaches targeted to the ER for the treatment of hormone-responsive breast cancer.


Asunto(s)
Adenoviridae/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/virología , Estradiol/farmacología , Regulación Neoplásica de la Expresión Génica , Receptores de Estrógenos/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , División Celular/genética , ADN sin Sentido , Estradiol/metabolismo , Femenino , Genes Dominantes , Genes myc , Vectores Genéticos/genética , Humanos , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transcripción Genética , Células Tumorales Cultivadas
10.
Mol Cell Endocrinol ; 149(1-2): 93-105, 1999 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-10375022

RESUMEN

Estrogen receptor (ER)-negative breast carcinomas are often difficult to treat as they do not respond to hormone therapy. In an attempt to determine if expressing the human estrogen receptor in an ectopic manner could restore the hormone responsiveness of these cells, we have expressed the human ER in ER-negative MDA-MB 231 breast cancer cells using a recombinant adenovirus gene delivery system that allows high level expression of ER in essentially all cells. In these cells, the ER was correctly translated, had a wild type hormone binding affinity (Kd = 0.6 nM), bound well to estrogen response element-containing DNA, and showed an activation pattern of estrogen response element-reporter gene activity by estrogen and antiestrogens very similar to that observed in MCF-7 breast cancer cells containing endogenous ER (stimulation by estrogen, no stimulation by the antiestrogens trans-hydroxytamoxifen or ICI 164384, and blockade of estradiol stimulation by trans-hydroxytamoxifen or ICI 164384). Intriguingly, estradiol stimulation of these cells was also able to induce expression of pS2, an estrogen regulated gene considered to be a favorable prognostic marker for endocrine therapy in ER-positive breast cancer cells. Expression of the ER had no effect by itself on the proliferation rate of MDA-MB 231 cells. However, treatment of the ER-containing cells with estradiol or with the pure antiestrogen ICI 164 384 suppressed proliferation of the cells while the antiestrogen trans-hydroxytamoxifen had little effect on proliferation; and cotreatment with trans-hydroxytamoxifen reversed the estradiol- or ICI 164 384-evoked suppression of proliferation. To understand the mechanism underlying the inhibition of proliferation by estradiol, we examined the expression of several growth related endogenous genes. c-Myc protooncogene expression was strongly inhibited by treatment with estradiol as was expression of BRCA1 and BRCA2 genes, which is in agreement with their mitogenic-dependent expression, while expression of beta-actin, a housekeeping gene, was not affected by hormone treatment. Together, these data suggest that reexpressing the human ER in breast cancer cells that no longer express this protein renders them sensitive to hormone treatment. The ability of the antiestrogen ICI 164 384 to suppress the proliferation of ER-negative breast cancer cells that reexpress ER might be useful ultimately as an endocrine gene therapy approach for controlling the growth of ER-negative breast cancer cells. The application of recombinant adenoviruses expressing the human ER presents interesting features which might be used as a basis for designing more powerful and effective treatments for ER-negative breast cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Técnicas de Transferencia de Gen , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Receptores de Estrógenos/genética , Adenoviridae/genética , Secuencia de Bases , Neoplasias de la Mama/terapia , División Celular , Cartilla de ADN/genética , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos , Humanos , Cinética , Neoplasias Hormono-Dependientes/terapia , Alcamidas Poliinsaturadas , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas
11.
Mol Cell Biol ; 17(9): 5053-66, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9271383

RESUMEN

The rainbow trout estrogen receptor (rtER) is a positively autoregulated gene in liver cells. In a previous report, we showed that upregulation is mediated by an estrogen response element (ERE) located in the proximal promoter of the gene and that a half binding site for nuclear receptors (5'-TGACCT-3') located 15 bp upstream of the ERE is involved in the magnitude of the estrogen response. We now report that the human orphan receptor COUP-TF and a COUP-TF-like protein from trout liver are able to bind to the consensus half-site. When cotransfected with the rtER gene proximal promoter, COUP-TF had no regulatory functions on its own. Interestingly, COUP-TF enhanced rtER transactivation properties in the presence of estradiol in a dose-dependent manner when cotransfected with the rtER gene promoter. Unliganded retinoid receptor heterodimers had the same helper function as COUP-TF in the presence of estradiol but were switched to repressors when the ligand all-trans-retinoic acid was added. Mutation of the consensus half-site only slightly reduced COUP-TF helper function, suggesting that it actually results from a complex mechanism that probably involves both DNA binding of COUP-TF to the promoter and protein-protein interaction with another transcription factor bound to the promoter. Nevertheless, a DNA-binding-defective mutant of COUP-TF was also defective in ER helper function. Competition footprinting analysis suggested that COUP-TF actually establishes contacts with the consensus upstream half-site and the downstream ERE half-site that would form a DR-24-like response element. Interaction of COUP-TF with the DR-24 element was confirmed in footprinting assays by using nuclear extracts from Saccharomyces cerevisiae expressing COUP-TF. Finally, interaction of COUP-TF with mutants of the rtER gene promoter showed that COUP-TF recognizes the ERE when the upstream half-site is mutated. These data show that COUP-TF may activate transcription through interaction with other nuclear receptors. This cross-talk between liganded nuclear receptors and orphan receptors is likely to modulate the spectrum of action of a particular ligand-receptor complex and may participate in the cell-type specificity of the ligand effect.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Homeostasis/genética , Receptores de Estrógenos/genética , Receptores de Glucocorticoides/fisiología , Receptores de Esteroides/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Células COS , Factor de Transcripción COUP I , Factor de Transcripción COUP II , Factores de Transcripción COUP , ADN/metabolismo , Huella de ADN , Humanos , Datos de Secuencia Molecular , Oncorhynchus mykiss , Regiones Promotoras Genéticas , Unión Proteica , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide , Tilapia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
12.
Mol Endocrinol ; 11(9): 1375-86, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9259327

RESUMEN

The estrogen receptor (ER) belongs to a large family of nuclear receptors, many of whose members function as ligand-dependent transcriptional activators. The mechanism by which the receptor is converted from an inactive into an activated state is not yet completely understood. To investigate the kind of changes in receptor conformation and interactions that are involved in this activation, we have used the wild type ER and a set of constitutively active ER point mutants that show from 20% to nearly 100% activity in the absence of estrogen. These mutants are of particular interest as they could mimic, in the absence of ligand, the activated state of the wild type receptor. We have analyzed several transcriptional steps that could be involved in the activation: the ability of these receptors 1) to interact with several coactivators (steroid receptor coactivator-1, SRC-1; transcription intermediary factor-1, TIF-1; and estrogen receptor-associated protein 140, ERAP 140) and with members of the preinitiation complex [TATA box-binding protein (TBP), transcription factor IIB (TFIIB)]; 2) to exhibit conformational changes revealed by proteolytic digest patterns similar to those observed for the wild type hormone-occupied ER; and 3) to bend estrogen response element-containing DNA, which is thought to be one of the important phenomena triggering transcriptional activation. Our results demonstrate that the interaction of these mutant receptors with coactivators is likely to be one of the features of the activated step, as the mutant receptors interacted with some coactivators in a ligand-independent manner in proportion to their extent of constitutive activity. However, the different degrees of ligand-independent interaction of the mutant ERs with the three coactivators suggest that SRC-1, TIF-1, and ERAP 140 may play different roles in receptor activity. Limited proteolytic digest experiments reveal that the activated state of the receptor corresponds to a particular conformation of the receptor, which is fully observed with the mutant ER showing the highest activity in the absence of estrogen. Finally, it appears that in inactive or active states, the receptor exhibits distinctly different DNA-bending abilities. Addition of estradiol is able to modify the bending ability of only the wild type receptor, whereas estradiol has no influence on the constitutive receptors, which exhibited the same bending ability as that observed for the ligand-occupied wild type receptor. These data document that the ER undergoes major changes in its conformation and also in its functional properties when it is turned from an inactive into an active state and that mutational changes in the ER protein that result in constitutive, hormone-independent activation mimic many of the changes in ER properties that are normally under hormone regulation.


Asunto(s)
Receptores de Estrógenos/química , Receptores de Estrógenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , ADN/química , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Electroforesis/métodos , Estradiol/metabolismo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Histona Acetiltransferasas , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Coactivador 1 de Receptor Nuclear , Proteína de Interacción con Receptores Nucleares 1 , Conformación Proteica , Desnaturalización Proteica , Receptores de Estrógenos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Proteína de Unión a TATA-Box , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Factor de Transcripción TFIIB , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
13.
Mol Endocrinol ; 10(11): 1388-98, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8923465

RESUMEN

To better understand structure-activity relationships in the human estrogen receptor (ER), we examined the role of tyrosine 537 in the transcriptional response of the receptor, since this residue is close to a region of the hormone-binding domain shown previously to be important in hormone-dependent transcriptional activity and because this amino acid has been proposed to be a tyrosine kinase phosphorylation site important in the activity of the ER. We substituted five amino acids at this position (alanine, phenylalanine, glutamic acid, lysine, or serine) and screened these mutants for their biological activities in the presence and absence of estradiol. Two of the ER mutants, Y537A and Y537S, displayed estrogen-independent constitutive activity that was approximately 20% or 100%, respectively, of the activity of the wild type receptor with estradiol, when assessed in two different cell backgrounds using three different estrogen-responsive promoters. In some circumstances, the Y537E and Y537K proteins also exhibited some low level of constitutive activity. The constitutive activity of the mutants, as well as their activity in the presence of E2, was fully suppressed by antiestrogen. The extent of interaction of the constitutively active ERs with the steroid receptor coactivator-1 (SRC-1) closely parallel the magnitude of transcriptional activity of the receptor. Whereas wild type ER showed interaction with SRC-1 only in the presence of estrogen, Y537A and Y537S ER showed moderate or full interaction in the absence of ligand, an interaction that was blocked by antiestrogen, and the magnitude of interaction was increased to or remained at 100% upon estradiol treatment, implying that the ability of an ER to associate with SRC-1 is a good indicator of a transcriptionally active conformational state of the receptor. Our findings indicate that tyrosine 537 is in a region important in the ligand regulation of ER transcriptional activity and that the presence of certain amino acids at this position can shift ER into a conformation that is active even without ligand. However, tyrosine is not required at this site for estrogen binding or transcriptional response to estrogen in the systems investigated. Our findings, interpreted in light of the recently published x-ray crystal structure of the ligand-binding domains of three related receptors of the nuclear receptor superfamily, suggest that some of the amino acid substitutions introduced at position 537 may facilitate the shift of helix 12 of the ER into an active conformation and/or allow for differential stabilization of the receptor in its active form.


Asunto(s)
Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Tirosina/genética , Alanina/genética , Alanina/metabolismo , Secuencia de Aminoácidos , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células CHO/efectos de los fármacos , Células CHO/metabolismo , Cricetinae , Estradiol/metabolismo , Estradiol/farmacología , Histona Acetiltransferasas , Humanos , Datos de Secuencia Molecular , Mutación , Coactivador 1 de Receptor Nuclear , Fenotipo , Fosforilación , Receptores de Estrógenos/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/genética , Serina/metabolismo , Relación Estructura-Actividad , Factores de Transcripción/metabolismo , Transcripción Genética , Células Tumorales Cultivadas , Tirosina/metabolismo
14.
Mol Endocrinol ; 10(9): 1116-26, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8885246

RESUMEN

The human estrogen receptor (hER) is expressed in breast cancer MCF-7 cells and plays a major role in tumorigenic processes. In this report, we demonstrate that MCF-7-specific factors can cooperate with the hER to increase its transactivation activity. We previously demonstrated that the rainbow trout ER (rtER) gene is up-regulated by the rtER protein itself, through an enhancer that contains an imperfect estrogen-responsive element (FP1 area). By performing footprinting experiments, we have delineated two other regulatory regions (FP2 and FP3 areas) in the 0.2-kb enhancer. We show, by transient transfections, that hER poorly transactivates this enhancer in CHO-K1 and Ishikawa cells whereas, in MCF-7 cells, transcriptional activation occurs at a level about 20-fold higher than when the enhancer estrogen-responsive element (FP1) is the only regulatory region included in the reporter gene. These results indicate that areas other than FP1 are important regulatory sites of this enhancer. Site-directed mutagenesis demonstrated that the FP1 area is absolutely necessary for induction by estradiol as well as for basal activity of this enhancer in MCF-7 cells. Gel shift experiments showed that MCF-7 cells contain a factor that binds to the FP3 area and is poorly expressed in all other tested cell lines. As suggested by site-directed mutagenesis and deletion experiments, this FP3-binding protein may enhance the hER transactivation ability in MCF-7 cells. These data reinforce the idea that cell-specific transcription factors cooperate with steroid receptors to achieve maximal induction of hormone-responsive genes.


Asunto(s)
Neoplasias de la Mama/genética , Elementos de Facilitación Genéticos/efectos de los fármacos , Oncorhynchus mykiss/genética , Receptores de Estrógenos/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células CHO/metabolismo , Cricetinae , Huella de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasa I/metabolismo , Estradiol/farmacología , Humanos , Hígado/química , Hígado/metabolismo , Datos de Secuencia Molecular , Mutación , Regiones Promotoras Genéticas , Receptores de Estrógenos/genética , Factores de Transcripción/farmacología , Transcripción Genética , Transfección , Células Tumorales Cultivadas
15.
Mol Cell Endocrinol ; 121(2): 153-63, 1996 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-8892316

RESUMEN

We have prepared an ovine pituitary cDNA library, isolated a clone containing the full-coding sequence of estrogen receptor (ER) cDNA, and determined its primary structure. This cDNA encodes a protein of 596 amino acids which shows great homology to other mammalian ER sequences, the highest degree being 95% with the porcine receptor. Northern blot analysis of ovine pituitary RNA revealed a 6.3 kb transcript. This receptor was showed to bind a consensus ERE and to be transcriptionally activated by E2. Studies investigating the pattern of expression of the ovine ER mRNA were also carried out, using the reverse transcription/PCR technique. Expression of ER mRNA was analyzed in ram pituitary and hypothalamus after contrasted light regimen and castration. Results showed that the light regimen had no effect on ER mRNA expression whereas castration induced a slight (approximately 20%) but significant increase of ER mRNA expression at both the hypothalamic (P < 0.05) and pituitary (P < 0.01) levels, indicating a negative regulation of ER gene expression by testicular steroids. Since we have previously shown no variations in ER protein levels after castration, data suggest the activation of a complex pattern including both transcriptional and post-transcriptional regulatory mechanisms in the ram hypothalamo-pituitary axis.


Asunto(s)
ADN Complementario/genética , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Hipófisis/metabolismo , Receptores de Estrógenos/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario/aislamiento & purificación , Datos de Secuencia Molecular , Receptores de Estrógenos/metabolismo , Alineación de Secuencia , Ovinos
16.
Gene ; 166(2): 243-7, 1995 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-8543169

RESUMEN

The estrogen receptor (ER)-encoding gene (ER) regulates many genes implicated in the reproductive functions. Moreover, rainbow trout ER (rtER) is itself up-regulated by its own product. We have used Northern blot, RNase protection, primer extension and reverse transcription-polymerase chain reaction (RT-PCR) to study the position of the rtER mRNA transcription start point (tsp) in liver. This analysis has revealed the presence of a tsp positioned at the beginning of the cloned rtER cDNA. Functionality of this tsp was tested in transient transfections in CHO-K1 cells. The characterization of the rtER 5' untranslated region (UTR) showed that two transcripts exist in liver which differ in their 5'-UTR. The first one is 100% homologous to the cloned rtER cDNA sequence. The other one contains a 41-bp insertion. The isolation and sequencing of the first intron showed that this insertion arises from alternative splicing, due to the use of a splicing site internal to the first intron.


Asunto(s)
Oncorhynchus mykiss/genética , Receptores de Estrógenos/genética , Empalme Alternativo , Animales , Secuencia de Bases , Células CHO , Cricetinae , Datos de Secuencia Molecular , ARN Mensajero/genética , Transcripción Genética , Transfección
17.
J Mol Endocrinol ; 15(1): 37-47, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8546812

RESUMEN

We previously reported that the expression of the rainbow trout estrogen receptor (rtER) gene is markedly increased by estradiol (E2). In this paper, we have used transient transfection assays with reporter plasmids expressing chloramphenicol acetyl transferase (CAT), linked to 5' flanking regions of the rtER gene promoter, to identify cis-elements responsible for E2 inducibility. Deletion analysis localized an estrogen-responsive element (ERE), at position +242, with one mutation on the first base compared with the consensus sequence. This element confers estrogen responsiveness to CAT reporter linked to both the herpes simplex virus thymidine kinase promoter and the homologous rtER promoter. Moreover, using a 0.2 kb fragment of the rtER promoter encompassing the ERE and the rtER DNA binding domain obtained from a bacterial expression system, DNase I footprinting experiments demonstrated a specific protection covering 20 bp (+240/+260) containing the ERE sequence. Based on these studies, we believe that this ERE sequence, identified in the rtER gene promoter, may be a major cis-acting element involved in the regulation of the gene by estrogen.


Asunto(s)
Estrógenos/fisiología , Regulación de la Expresión Génica/fisiología , Oncorhynchus mykiss/genética , Receptores de Estrógenos/genética , Animales , Secuencia de Bases , Células CHO , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Clonación Molecular , Cricetinae , ADN Recombinante , Datos de Secuencia Molecular , Mutación , Regiones Promotoras Genéticas , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Transcripción Genética , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...