Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Biol Chem ; 291(1): 342-54, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26504077

RESUMEN

Most of the steps in, and many of the factors contributing to, glucocorticoid receptor (GR)-regulated gene induction are currently unknown. A competition assay, based on a validated chemical kinetic model of steroid hormone action, is now used to identify two new factors (BRD4 and negative elongation factor (NELF)-E) and to define their sites and mechanisms of action. BRD4 is a kinase involved in numerous initial steps of gene induction. Consistent with its complicated biochemistry, BRD4 is shown to alter both the maximal activity (Amax) and the steroid concentration required for half-maximal induction (EC50) of GR-mediated gene expression by acting at a minimum of three different kinetically defined steps. The action at two of these steps is dependent on BRD4 concentration, whereas the third step requires the association of BRD4 with P-TEFb. BRD4 is also found to bind to NELF-E, a component of the NELF complex. Unexpectedly, NELF-E modifies GR induction in a manner that is independent of the NELF complex. Several of the kinetically defined steps of BRD4 in this study are proposed to be related to its known biochemical actions. However, novel actions of BRD4 and of NELF-E in GR-controlled gene induction have been uncovered. The model-based competition assay is also unique in being able to order, for the first time, the sites of action of the various reaction components: GR < Cdk9 < BRD4 ≤ induced gene < NELF-E. This ability to order factor actions will assist efforts to reduce the side effects of steroid treatments.


Asunto(s)
Proteínas Nucleares/metabolismo , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Animales , Unión Competitiva , Proteínas de Ciclo Celular , Quinasa 9 Dependiente de la Ciclina/metabolismo , Células HeLa , Humanos , Cinética , Proteínas Mutantes/metabolismo , Mutación , Coactivador 2 del Receptor Nuclear/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Unión Proteica , Ratas
2.
Biochemistry ; 53(11): 1753-67, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24559102

RESUMEN

A gene induction competition assay has recently uncovered new inhibitory activities of two transcriptional cofactors, NELF-A and NELF-B, in glucocorticoid-regulated transactivation. NELF-A and -B are also components of the NELF complex, which participates in RNA polymerase II pausing shortly after the initiation of gene transcription. We therefore asked if cofactors (Cdk9 and ELL) best known to affect paused polymerase could reverse the effects of NELF-A and -B. Unexpectedly, Cdk9 and ELL augmented, rather than prevented, the effects of NELF-A and -B. Furthermore, Cdk9 actions are not blocked either by Ckd9 inhibitors (DRB or flavopiridol) or by two Cdk9 mutants defective in kinase activity. The mode and site of action of NELF-A and -B mutants with an altered NELF domain are similarly affected by wild-type and kinase-dead Cdk9. We conclude that Cdk9 is a new modulator of GR action, that Ckd9 and ELL have novel activities in GR-regulated gene expression, that NELF-A and -B can act separately from the NELF complex, and that Cdk9 possesses activities that are independent of Cdk9 kinase activity. Finally, the competition assay has succeeded in ordering the site of action of several cofactors of GR transactivation. Extension of this methodology should be helpful in determining the site and mode of action of numerous additional cofactors and in reducing unwanted side effects.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Receptores de Glucocorticoides/fisiología , Activación Transcripcional/genética , Animales , Células COS , Chlorocebus aethiops , Humanos , Mutación/genética , Ratas , Receptores de Glucocorticoides/antagonistas & inhibidores , Factores de Transcripción/fisiología
3.
J Biol Chem ; 285(41): 31100-6, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20705611

RESUMEN

Constitutive activation of NFκB in estrogen receptor (ER)-positive breast cancer is associated with tumor recurrence and development of anti-estrogen resistance. Furthermore, a gene expression signature containing common targets for ER and NFκB has been identified and found to be associated with the more aggressive luminal B intrinsic subtype of ER-positive breast tumors. Here, we describe a novel mechanism by which ER and NFκB cooperate to up-regulate expression of one important gene from this signature, ABCG2, which encodes a transporter protein associated with the development of drug-resistant breast cancer. We and others have confirmed that this gene is regulated primarily by estrogen in an ER- and estrogen response element (ERE)-dependent manner. We found that whereas proinflammatory cytokines have little effect on this gene in the absence of 17ß-estradiol, they can potentiate ER activity in an NFκB-dependent manner. ER allows the NFκB family member p65 to access a latent NFκB response element located near the ERE in the gene promoter. NFκB recruitment to the gene is, in turn, required to stabilize ER occupancy at the functional ERE. The result of this cooperative binding of ER and p65 at adjacent response elements leads to a major increase in both ABCG2 mRNA and protein expression. These findings indicate that estrogen and inflammatory factors can modify each other's activity through modulation of transcription factor accessibility and/or occupancy at adjacent response elements. This novel transcriptional mechanism could have important implications in breast cancer, where both inflammation and estrogen can promote cancer progression.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/biosíntesis , Neoplasias de la Mama/metabolismo , Citocinas/metabolismo , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Receptores de Estrógenos/metabolismo , Elementos de Respuesta , Factor de Transcripción ReIA/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Neoplasias de la Mama/genética , Línea Celular Tumoral , Citocinas/genética , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Humanos , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Estrógenos/genética , Factor de Transcripción ReIA/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
4.
Endocrinology ; 149(12): 6272-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18703630

RESUMEN

Inflammatory mediators, such as cytokines and prostaglandins, play a fundamental role in estrogen-dependent breast cancer through their ability to up-regulate aromatase expression and subsequent local production of estrogens in the breast. To study the link between estrogens and inflammation further, we examined the regulation of prostaglandin E synthase (PTGES), a key enzyme in the production of prostaglandin E2. We found that 17beta-estradiol (E2) rapidly and robustly up-regulates PTGES mRNA and protein levels in estrogen receptor (ER)-positive breast cancer cells through ER recruitment to an essential estrogen response element located in the 5' flanking region of the PTGES gene. PTGES is also up-regulated by the proinflammatory cytokines TNFalpha or IL-1beta. Surprisingly, the combination of E2 and cytokines leads to a synergistic up-regulation of PTGES in an ER and nuclear factor-kappaB (NFkappaB)-dependent manner. This is in contrast to the mutual transrepression between ER and NFkappaB that has been well characterized in other cell types. Furthermore, we found enhanced recruitment of ERalpha as well as the NFkappaB family member, p65, to the PTGES estrogen response element by the combination of E2 and TNFalpha compared with either E2 or TNFalpha alone. The synergistic up-regulation of PTGES may result in enhanced prostaglandin E2 production, which in turn may further enhance aromatase expression and production of local estrogens. Our findings suggest that a finely tuned positive feedback mechanism between estrogens and inflammatory factors may exist in the breast and contribute to hormone-dependent breast cancer growth and progression.


Asunto(s)
Citocinas/farmacología , Estradiol/farmacología , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Sinergismo Farmacológico , Humanos , Interleucina-1beta/farmacología , Prostaglandina-E Sintasas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/farmacología
5.
Mol Endocrinol ; 28(7): 1194-206, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24850414

RESUMEN

Glucocorticoid steroids affect almost every type of tissue and thus are widely used to treat a variety of human pathological conditions. However, the severity of numerous side effects limits the frequency and duration of glucocorticoid treatments. Of the numerous approaches to control off-target responses to glucocorticoids, small molecules and pharmaceuticals offer several advantages. Here we describe a new, extended high-throughput screen in intact cells to identify small molecule modulators of dexamethasone-induced glucocorticoid receptor (GR) transcriptional activity. The novelty of this assay is that it monitors changes in both GR maximal activity (A(max)) and EC(50) (the position of the dexamethasone dose-response curve). Upon screening 1280 chemicals, 10 with the greatest changes in the absolute value of A(max) or EC(50) were selected for further examination. Qualitatively identical behaviors for 60% to 90% of the chemicals were observed in a completely different system, suggesting that other systems will be similarly affected by these chemicals. Additional analysis of the 10 chemicals in a recently described competition assay determined their kinetically defined mechanism and site of action. Some chemicals had similar mechanisms of action despite divergent effects on the level of the GR-induced product. These combined assays offer a straightforward method of identifying numerous new pharmaceuticals that can alter GR transactivation in ways that could be clinically useful.


Asunto(s)
Dexametasona/efectos adversos , Glucocorticoides/efectos adversos , Ensayos Analíticos de Alto Rendimiento/métodos , Receptores de Glucocorticoides/metabolismo , Activación Transcripcional/efectos de los fármacos , Línea Celular , Dexametasona/farmacología , Glucocorticoides/farmacología , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Receptores de Glucocorticoides/genética , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
6.
Mol Cell Biol ; 32(2): 569-75, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22083956

RESUMEN

Estrogen receptor (ER) and NF-κB are transcription factors with profound effects on breast cancer cell proliferation and survival. While many studies demonstrate that ER and NF-κB can repress each other, we previously identified a gene signature that is synergistically upregulated by these two factors in more aggressive luminal B breast tumors. Herein, we examine a novel mechanism of cross talk between ER and NF-κB that results in the upregulation of the antiapoptotic gene BIRC3 (also known as cIAP2). We demonstrate that NF-κB, acting through two response elements, is required for ER recruitment to an adjacent estrogen response element (ERE) in the BIRC3 promoter. This effect is accompanied by a major increase in NF-κB-dependent histone acetylation around the ERE. Interestingly, CBP, a histone acetyltransferase previously implicated in repressive interactions between ER and NF-κB, plays a permissive role by promoting histone acetylation and ER recruitment, as well as enhanced expression of BIRC3. These findings suggest a new gene regulatory mechanism by which inflammation and NF-κB activation can influence ER recruitment to inherently inactive ER binding sites. This fine-tuning mechanism may explain how two factors that generally repress each other's activity may work together on certain genes to promote breast cancer cell survival and tumor progression.


Asunto(s)
Neoplasias de la Mama/genética , Proteína de Unión a CREB/metabolismo , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Proteínas Inhibidoras de la Apoptosis/genética , FN-kappa B/metabolismo , Receptores de Estrógenos/metabolismo , Acetilación , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Elementos de Respuesta , Ubiquitina-Proteína Ligasas
7.
Cancer Res ; 69(23): 8918-25, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19920189

RESUMEN

Estrogen receptors (ER) and nuclear factor-kappaB (NF-kappaB) are known to play important roles in breast cancer, but these factors are generally thought to repress each other's activity. However, we have recently found that ER and NF-kappaB can also act together in a positive manner to synergistically increase gene transcription. To examine the extent of cross-talk between ER and NF-kappaB, a microarray study was conducted in which MCF-7 breast cancer cells were treated with 17beta-estradiol (E(2)), tumor necrosis factor alpha (TNFalpha), or both. Follow-up studies with an ER antagonist and NF-kappaB inhibitors show that cross-talk between E(2) and TNFalpha is mediated by these two factors. We find that although transrepression between ER and NF-kappaB does occur, positive cross-talk is more prominent with three gene-specific patterns of regulation: (a) TNFalpha enhances E(2) action on approximately 30% of E(2)-upregulated genes; (b) E(2) enhances TNFalpha activity on approximately 15% of TNFalpha-upregulated genes; and (c) E(2) + TNFalpha causes a more than additive upregulation of approximately 60 genes. Consistent with their prosurvival roles, ER and NF-kappaB and their target gene, BIRC3, are involved in protecting breast cancer cells against apoptosis. Furthermore, genes positively regulated by E(2) + TNFalpha are clinically relevant because they are enriched in luminal B breast tumors and their expression profiles can distinguish a cohort of patients with poor outcome following endocrine treatment. Taken together, our findings suggest that positive cross-talk between ER and NF-kappaB is more extensive than anticipated and that these factors may act together to promote survival of breast cancer cells and progression to a more aggressive phenotype.


Asunto(s)
Neoplasias de la Mama/metabolismo , FN-kappa B/metabolismo , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Estradiol/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptor Cross-Talk , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA