Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nat Genet ; 38(11): 1289-97, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17013392

RESUMEN

The estrogen receptor is the master transcriptional regulator of breast cancer phenotype and the archetype of a molecular therapeutic target. We mapped all estrogen receptor and RNA polymerase II binding sites on a genome-wide scale, identifying the authentic cis binding sites and target genes, in breast cancer cells. Combining this unique resource with gene expression data demonstrates distinct temporal mechanisms of estrogen-mediated gene regulation, particularly in the case of estrogen-suppressed genes. Furthermore, this resource has allowed the identification of cis-regulatory sites in previously unexplored regions of the genome and the cooperating transcription factors underlying estrogen signaling in breast cancer.


Asunto(s)
Genoma Humano , Receptores de Estrógenos/metabolismo , Elementos de Respuesta , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/genética , Neoplasias de la Mama/genética , Células Cultivadas , Mapeo Cromosómico/métodos , Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Expresión Génica , Regulación de la Expresión Génica , Humanos , Análisis por Micromatrices/métodos , Proteínas Nucleares/metabolismo , Proteína de Interacción con Receptores Nucleares 1 , Elementos de Respuesta/fisiología , Factores de Transcripción/fisiología , Sitio de Iniciación de la Transcripción
2.
Nature ; 456(7222): 663-6, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19005469

RESUMEN

Crosstalk between the oestrogen receptor (ER) and ERBB2/HER-2 pathways has long been implicated in breast cancer aetiology and drug response, yet no direct connection at a transcriptional level has been shown. Here we show that oestrogen-ER and tamoxifen-ER complexes directly repress ERBB2 transcription by means of a cis-regulatory element within the ERBB2 gene in human cell lines. We implicate the paired box 2 gene product (PAX2), in a previously unrecognized role, as a crucial mediator of ER repression of ERBB2 by the anti-cancer drug tamoxifen. We show that PAX2 and the ER co-activator AIB-1/SRC-3 compete for binding and regulation of ERBB2 transcription, the outcome of which determines tamoxifen response in breast cancer cells. The repression of ERBB2 by ER-PAX2 links these two breast cancer subtypes and suggests that aggressive ERBB2-positive tumours can originate from ER-positive luminal tumours by circumventing this repressive mechanism. These data provide mechanistic insight into the molecular basis of endocrine resistance in breast cancer.


Asunto(s)
Genes erbB-2/genética , Factor de Transcripción PAX2/metabolismo , Receptor ErbB-2/genética , Receptores de Estrógenos/metabolismo , Tamoxifeno/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Resistencia a Antineoplásicos/genética , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Histona Acetiltransferasas , Humanos , Coactivador 3 de Receptor Nuclear , Factor de Transcripción PAX2/deficiencia , Factor de Transcripción PAX2/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas Represoras/metabolismo , Tamoxifeno/metabolismo , Transactivadores
3.
Chem Biol ; 11(2): 273-81, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15123288

RESUMEN

Ligand-dependent nuclear hormone receptor (NR) signaling requires direct interaction between NR and the steroid receptor coactivators (SRC). Herein we utilize a library of SRC2 peptidomimetics to select for specific inhibitors of the interaction of SRC2 with the two estrogen receptor (ER) isoforms, ERalpha and ERbeta, in the presence of three different ligands: 17beta-estradiol, diethylstilbesterol, and genistein. The pattern of inhibitor selectivity for each ER isoform varied depending upon which ligand was present, thus demonstrating that the ligands exert unique allosteric effects upon the surface of the SRC binding pocket. Several of the lead compounds are highly (>100-fold) selective for blocking the binding of SRC2 to ERalpha, in preference to ERbeta, in the presence of one ligand and therefore may prove useful for decoupling ERbeta signaling from ERalpha signaling.


Asunto(s)
Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Esteroides/antagonistas & inhibidores , Animales , Sitios de Unión , Biomimética , Dietilestilbestrol/farmacología , Estrógenos/farmacología , Genisteína/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Isoformas de Proteínas/antagonistas & inhibidores , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/química , Receptores de Esteroides/química , Relación Estructura-Actividad
4.
Mol Cell Biol ; 30(8): 1866-77, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20123965

RESUMEN

An analysis of mRNA expression in T47D breast cancer cells treated with the synthetic progestin R5020 revealed a subset of progesterone receptor (PR) target genes that are enriched for E2F binding sites. Following up on this observation, we determined that PR-B acts in both direct and indirect manners to positively upregulate E2F1 expression in T47D cells. The direct effects of PR on E2F1 expression were confirmed by chromatin immunoprecipitation (ChIP) analysis, which indicated that the agonist-bound receptor was recruited to several enhancer elements proximal to the E2F1 transcript. However, we also noted that cycloheximide partially inhibits R5020 induction of E2F1 expression, indicating that the ligand-dependent actions of PR on this gene may involve additional indirect regulatory pathways. In support of this hypothesis, we demonstrated that treatment with R5020 significantly increases both hyperphosphorylation of Rb and recruitment of E2F1 to its own promoter, thus activating a positive feedback loop that further amplifies its transcription. Furthermore, we established that PR-mediated induction of Krüppel-like factor 15 (KLF15), which can bind to GC-rich DNA within the E2F1 promoter, is required for maximal induction of E2F1 expression by progestins. Taken together, these results suggest a new paradigm for multimodal regulation of target gene expression by PR.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Progestinas/farmacología , Receptores de Progesterona/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Factor de Transcripción E2F1/genética , Femenino , Perfilación de la Expresión Génica , Genes Reporteros , Humanos , Factores de Transcripción de Tipo Kruppel , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nucleares , Análisis de Secuencia por Matrices de Oligonucleótidos , Plicamicina/análogos & derivados , Plicamicina/farmacología , Promegestona/farmacología , Regiones Promotoras Genéticas , Receptores de Progesterona/genética , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
6.
Genes Dev ; 20(18): 2513-26, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16980581

RESUMEN

Estrogen stimulates the proliferation of the most common type of human breast cancer that expresses estrogen receptor alpha (ERalpha) through the activation of the cyclin D1 (CCND1) oncogene. However, our knowledge of ERalpha transcriptional mechanisms remains limited. Hence, it is still elusive why ERalpha ectopically expressed in ER-negative breast cancer cells (BCC) is functional on ectopic reporter constructs but lacks activity on many endogenous target genes, including CCND1. Here, we show that estradiol (E2) stimulation of CCND1 expression in BCC depends on a novel cell-type-specific enhancer downstream from the CCND1 coding region, which is the primary ERalpha recruitment site in estrogen-responsive cells. The pioneer factor FoxA1 is specifically required for the active chromatin state of this enhancer and as such is crucial for both CCND1 expression and subsequent cell cycle progression. Interestingly, even in BCC, CCND1 levels and proliferation are tightly controlled by E2 through the establishment of a negative feedforward loop involving the induction of NFIC, a putative tumor suppressor capable of directly repressing CCND1 transcription. Taken together, our results reveal an estrogen-regulated combinatorial network including cell-specific cis- and trans-regulators of CCND1 expression where ERalpha collaborates with other transcription factors associated with the ER-positive breast cancer phenotype, including FoxA1 and NFIC.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclina D1/genética , Estradiol/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , ADN Polimerasa II/metabolismo , Elementos de Facilitación Genéticos , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , Proteínas con Dominio LIM , Proteínas de Microfilamentos/genética , Modelos Biológicos , Factores de Transcripción NFI/genética , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Transcripción Genética/efectos de los fármacos
7.
Cell ; 122(1): 33-43, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16009131

RESUMEN

Estrogen plays an essential physiologic role in reproduction and a pathologic one in breast cancer. The completion of the human genome has allowed the identification of the expressed regions of protein-coding genes; however, little is known concerning the organization of their cis-regulatory elements. We have mapped the association of the estrogen receptor (ER) with the complete nonrepetitive sequence of human chromosomes 21 and 22 by combining chromatin immunoprecipitation (ChIP) with tiled microarrays. ER binds selectively to a limited number of sites, the majority of which are distant from the transcription start sites of regulated genes. The unbiased sequence interrogation of the genuine chromatin binding sites suggests that direct ER binding requires the presence of Forkhead factor binding in close proximity. Furthermore, knockdown of FoxA1 expression blocks the association of ER with chromatin and estrogen-induced gene expression demonstrating the necessity of FoxA1 in mediating an estrogen response in breast cancer cells.


Asunto(s)
Mapeo Cromosómico , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Estrógenos/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Inmunoprecipitación de Cromatina/métodos , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 22/genética , Proteínas de Unión al ADN/genética , Estrógenos/genética , Estrógenos/metabolismo , Regulación de la Expresión Génica/genética , Factor Nuclear 3-alfa del Hepatocito , Humanos , Ratones , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Unión Proteica/genética , Receptores de Estrógenos/genética , Factores de Transcripción/genética
8.
J Am Chem Soc ; 125(23): 6852-3, 2003 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-12783522

RESUMEN

Nuclear hormone receptor (NR) signaling, currently a therapeutic target in multiple diseases, involves an ordered series of protein interactions to regulate transcription in response to changing hormone levels. Later steps in the process of ligand-dependent signaling are driven by a highly conserved interaction between the NRs and the steroid receptor coactivators (SRCs) that is effected by a conserved interaction motif (L1XXL2L3), known as an NR box. Using computational design and combinatorial chemistry, we have produced novel alpha-helical proteomimetics of the second NR box of SRC2 that exploit structural differences between human estrogen receptor alpha (hERalpha), human estrogen receptor beta (hERbeta), and human thyroid hormone receptor beta (hTRbeta). The resulting library sequentially replaced each leucine with non-natural side chains. Screening this library using a quantitative competition assay revealed compounds that selectively inhibit the interaction of SRC2-2 with each individual NR in preference to its interaction with the other NR. This approach generated highly selective compounds from one that had no specificity for a particular family member. These compounds represent the first family-member-selective competitive inhibitors of the protein interactions of transcription factors.


Asunto(s)
Materiales Biomiméticos/farmacología , Péptidos/farmacología , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Materiales Biomiméticos/química , Receptor alfa de Estrógeno , Coactivador 2 del Receptor Nuclear , Péptidos/síntesis química , Péptidos/química , Receptores de Estrógenos/química , Receptores de Hormona Tiroidea/química , Receptores beta de Hormona Tiroidea , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA