Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 288(44): 31930-6, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24030830

RESUMEN

The circadian clock enables organisms to adjust to daily environmental changes and synchronize multiple molecular, biochemical, physiological, and behavioral processes accordingly. In mammalian clock work, Bmal1 is the most important core clock gene, which works with another core clock gene Clock to drive the expression of other clock genes and clock-controlled genes. However, the regulation of Bmal1 has not been fully understood. This work was aimed at identifying the positive regulator(s) of Bmal1 transcription. A series of 5' deletion reporter constructs was generated, and binding site mutations of mouse Bmal1 promoter fragments were cloned into pGL3-basic and pGL3(R2.1)-basic plasmids and transfected into NIH 3T3 cells. Luciferase activity was either measured 48 h after transfection or recorded for 4 days after serum shock. DNA affinity precipitation assay was used to detect the transcription factors binding to Bmal1 promoter. Small interfering RNA against nuclear factor Y, subunit A (NF-YA) and dominant negative NF-YA were employed to study the role of NF-Y in Bmal1 transcription regulation. Deletion and mutation analyses identified two clusters of CCAAT/GC-boxes at the proximal region of Bmal1 promoter as the activating cis-elements. Bmal1 promoter activity was up-regulated by NF-Y and/or Sp1 and repressed by dominant negative NF-YA or siRNA against NF-YA. The activation of Bmal1 promoter activity by NF-Y and Sp1 was inhibited by Rev-Erbα. DNA affinity precipitation assay showed that NF-Y and Sp1 bound to the two CCAAT/GC clusters of Bmal1 promoter. These results indicate that NF-Y is a functional activator of Bmal1 transcription and it cooperates with Sp1 and Rev-Erbα to generate the daily cycle of Bmal1 expression.


Asunto(s)
Factores de Transcripción ARNTL/biosíntesis , Factor de Unión a CCAAT/metabolismo , Relojes Circadianos/fisiología , Regulación de la Expresión Génica/fisiología , Elementos de Respuesta/fisiología , Transcripción Genética/fisiología , Factores de Transcripción ARNTL/genética , Animales , Factor de Unión a CCAAT/genética , Ratones , Células 3T3 NIH , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo
2.
J Biol Chem ; 287(37): 31280-97, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22815488

RESUMEN

Thyroid hormone receptor (TR) is a member of the nuclear receptor superfamily that shuttles between the cytosol and nucleus. The fine balance between nuclear import and export of TR has emerged as a critical control point for modulating thyroid hormone-responsive gene expression; however, sequence motifs of TR that mediate shuttling are not fully defined. Here, we characterized multiple signals that direct TR shuttling. Along with the known nuclear localization signal in the hinge domain, we identified a novel nuclear localization signal in the A/B domain of thyroid hormone receptor α1 that is absent in thyroid hormone receptor ß1 and inactive in the oncoprotein v-ErbA. Our prior studies showed that thyroid hormone receptor α1 exits the nucleus through two pathways, one dependent on the export factor CRM1 and the other CRM1-independent. Here, we identified three novel CRM1-independent nuclear export signal (NES) motifs in the ligand-binding domain as follows: a highly conserved NES in helix 12 (NES-H12) and two additional NES sequences spanning helix 3 and helix 6, respectively. Mutations predicted to disrupt the α-helical structure resulted in a significant decrease in NES-H12 activity. The high degree of conservation of helix 12 suggests that this region may function as a key NES in other nuclear receptors. Furthermore, our mutagenesis studies on NES-H12 suggest that altered shuttling of thyroid hormone receptor ß1 may be a contributing factor in resistance to thyroid hormone syndrome. Taken together, our findings provide a detailed mechanistic understanding of the multiple signals that work together to regulate TR shuttling and transcriptional activity, and they provide important insights into nuclear receptor function in general.


Asunto(s)
Señales de Localización Nuclear/metabolismo , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Secuencias de Aminoácidos , Animales , Células HeLa , Humanos , Mutación , Señales de Localización Nuclear/genética , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , Estructura Terciaria de Proteína , Ratas , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/genética
3.
Biochem Biophys Res Commun ; 413(3): 414-9, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21893042

RESUMEN

The control of energy homeostasis within the hypothalamus is under the regulated control of homeostatic hormones, nutrients and the expression of neuropeptides that alter feeding behavior. Elevated levels of palmitate, a predominant saturated fatty acid in diet and fatty acid biosynthesis, alter cellular function. For instance, a key mechanism involved in the development of insulin resistance is lipotoxicity, through increased circulating saturated fatty acids. Although many studies have begun to determine the underlying mechanisms of lipotoxicity in peripheral tissues, little is known about the effects of excess lipids in the brain. To determine these mechanisms we used an immortalized, clonal, hypothalamic cell line, mHypoE-44, to demonstrate that palmitate directly alters the expression of molecular clock components, by increasing Bmal1 and Clock, or by decreasing Per2, and Rev-erbα, their mRNA levels and altering their rhythmic period within individual neurons. We found that these neurons endogenously express the orexigenic neuropeptides NPY and AgRP, thus we determined that palmitate administration alters the mRNA expression of these neuropeptides as well. Palmitate treatment causes a significant increase in NPY mRNA levels and significantly alters the phase of rhythmic expression. We explored the link between AMPK and the expression of neuropeptide Y using the AMPK inhibitor compound C and the AMP analog AICAR. AMPK inhibition decreased NPY mRNA. AICAR also elevated basal NPY, but prevented the palmitate-mediated increase in NPY mRNA levels. We postulate that this palmitate-mediated increase in NPY and AgRP synthesis may initiate a detrimental positive feedback loop leading to increased energy consumption.


Asunto(s)
Proteínas CLOCK/genética , Ritmo Circadiano/genética , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Neuropéptido Y/genética , Palmitatos/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Factores de Transcripción ARNTL/genética , Animales , Células Cultivadas , Metabolismo Energético/genética , Productos del Gen rev/genética , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptidos/genética , Proteínas Oncogénicas v-erbA/genética , Orexinas , Palmitatos/farmacología , Proteínas Circadianas Period/genética , Fosforilación , Proteínas Quinasas/metabolismo , ARN Mensajero/biosíntesis
4.
Mol Biol Rep ; 38(2): 1137-44, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20571909

RESUMEN

Mutant forms of thyroid hormone receptor (TR) with dominant negative activity are frequently found in human hepatocellular carcinoma (HCC). Interestingly, the v-erbA oncogene, known to exert a dominant-negative effect on the expression of thyroid hormone (T3)-responsive genes, led to the development of HCC in a transgenic mouse model. Thus it is possible that the oncogenic activity of v-erbA in hepatocytes may be mediated by its dominant negative activity on T3-responsive genes. Microarray analysis was used to identify genes differentially expressed in murine hepatocytes in culture (AML12 cells) stably transfected with v-erbA and exposed to T3. The Affymetrix GeneChip Mouse Genome 430 2.0 array consisted of over 39,000 transcripts representing well-known genes. We have identified twenty T3-responsive genes that are negatively regulated by v-erbA at 3 h, and eighteen genes at 24 h, such as follistatin, activin ßC, thrombomodulin, Six1, Rasgrp3 and Ndrg2, as well as genes that are regulated by v-erbA only, such as angiopoietin 1 and Igfr2. We have identified T3 responsive genes that are dysregulated by v-erbA. These genes are known to be involved in carcinogenesis. Our studies may provide insight into the potential role of mutant forms of TR in the pathogenesis of HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes erbA , Neoplasias Hepáticas/metabolismo , Proteínas Oncogénicas v-erbA/genética , Receptores de Hormona Tiroidea/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Línea Celular , Genes Dominantes , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas , Proteínas Oncogénicas v-erbA/metabolismo , Factores de Tiempo
5.
Mol Cell Biol ; 17(10): 6131-8, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9315673

RESUMEN

Nuclear hormone receptors are ligand-regulated transcription factors that modulate gene expression in response to small, hydrophobic hormones, such as retinoic acid and thyroid hormone. The thyroid hormone and retinoic acid receptors typically repress transcription in the absence of hormone and activate it in the presence of hormone. Transcriptional repression is mediated, in part, through the ability of these receptors to physically associate with ancillary polypeptides called corepressors. We wished to understand the mechanism by which corepressors are recruited to unliganded nuclear hormone receptors and are released on the binding of hormone. We report here that an alpha-helical domain located at the thyroid hormone receptor C terminus appears to undergo a hormone-induced conformational change required for release of corepressor and that amino acid substitutions that abrogate this conformational change can impair or prevent corepressor release. In contrast, retinoid X receptors appear neither to undergo an equivalent conformational alteration in their C termini nor to release corepressor in response to cognate hormone, consistent with the distinct transcriptional regulatory properties displayed by this class of receptors.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Receptores de Ácido Retinoico/química , Receptores de Hormona Tiroidea/química , Proteínas Represoras/metabolismo , Factores de Transcripción/química , Tretinoina/metabolismo , Triyodotironina/metabolismo , Secuencia de Aminoácidos , Carboxipeptidasas , Catepsina A , Dimerización , Humanos , Datos de Secuencia Molecular , Co-Represor 2 de Receptor Nuclear , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , Mutación Puntual , Conformación Proteica , Receptores de Ácido Retinoico/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Receptores X Retinoide , Factores de Transcripción/metabolismo
6.
Oncogene ; 24(45): 6737-52, 2005 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-16007162

RESUMEN

The v-Erb A oncoprotein of avian erythroblastosis virus is derived from c-Erb A, a hormone-activated transcription factor. Notably, v-Erb A has sustained multiple mutations relative to c-Erb A and functions as a constitutive transcriptional repressor. We report here an analysis of the contributions of these different mutations to v-Erb A function. Our experiments demonstrate that two amino-acid differences between v-Erb A and c-Erb A, located in the 'I-box,' alter the dimerization properties of the viral protein, resulting in more stable homodimer formation, increased corepressor binding, and increased target gene repression. An additional amino-acid difference between v- and c-Erb A, located in helix 3 of the hormone binding domain, renders corepressor binding by the viral protein more resistant to release by thyroid hormone. Finally, we report that a C-terminal truncation in v-Erb A not only inhibits exchange of corepressor and coactivator, as previously noted, but also permits v-Erb A to recruit both SMRT and N-CoR corepressors, whereas c-Erb A is selective for N-CoR. The latter two mutations in v-Erb A also impair its ability to suppress c-Jun function in response to T3 hormone. We propose that the acquisition of oncogenic potential by the v-Erb A protein was a multistep process involving a series of mutations that alter the transcriptional repressive properties of the viral protein through multiple mechanisms.


Asunto(s)
Mutación , Proteínas Oncogénicas v-erbA/fisiología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Dimerización , Ensayo de Cambio de Movilidad Electroforética , Mutagénesis Sitio-Dirigida , Proteínas Oncogénicas v-erbA/genética , Receptores X Retinoide/metabolismo
7.
Mol Endocrinol ; 19(5): 1213-30, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15650025

RESUMEN

The retroviral v-ErbA oncoprotein is a highly mutated variant of the thyroid hormone receptor alpha (TRalpha), which is unable to bind T(3) and interferes with the action of TRalpha in mammalian and avian cancer cells. v-ErbA dominant-negative activity is attributed to competition with TRalpha for T(3)-responsive DNA elements and/or auxiliary factors involved in the transcriptional regulation of T(3)-responsive genes. However, competition models do not address the altered subcellular localization of v-ErbA and its possible implications in oncogenesis. Here, we report that v-ErbA dimerizes with TRalpha and the retinoid X receptor and sequesters a significant fraction of the two nuclear receptors in the cytoplasm. Recruitment of TRalpha to the cytoplasm by v-ErbA can be partially reversed in the presence of ligand and when chromatin is disrupted by the histone deacetylase inhibitor trichostatin A. These results define a new mode of action of v-ErbA and illustrate the importance of cellular compartmentalization in transcriptional regulation and oncogenesis.


Asunto(s)
Neoplasias/metabolismo , Proteínas Oncogénicas v-erbA/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Citoplasma/metabolismo , Dimerización , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Carioferinas/metabolismo , Ligandos , Ratones , Células 3T3 NIH , Proteínas Oncogénicas v-erbA/genética , Transporte de Proteínas/fisiología , Receptor beta X Retinoide/metabolismo , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Proteína Exportina 1
8.
Cancer Res ; 63(24): 8968-76, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695215

RESUMEN

Angiogenic factors are necessary for tumor proliferation and thus are attractive therapeutic targets. In this study, we have used engineered zinc finger protein (ZFP) transcription factors (TFs) to repress expression of vascular endothelial growth factor (VEGF)-A in human cancer cell lines. We create potent transcriptional repressors by fusing a designed ZFP targeted to the VEGF-A promoter with either the ligand-binding domain of thyroid hormone receptor alpha or its viral relative, vErbA. Moreover, this ZFP-vErbA repressor binds its intended target site in vivo and mediates the specific deacetylation of histones H3 and H4 at the targeted promoter, a result that emulates the natural repression mechanism of these domains. The potential therapeutic relevance of ZFP-mediated VEGF-A repression was addressed using the highly tumorigenic glioblastoma cell line U87MG. Despite the aberrant overexpression of VEGF-A in this cell line, engineered ZFP TFs were able to repress the expression of VEGF-A by >20-fold. The VEGF-A levels observed after ZFP TF-mediated repression were comparable to those of a nonangiogenic cancer line (U251MG), suggesting that the degree of repression obtained with the ZFP TF would be sufficient to suppress tumor angiogenesis. Thus, engineered ZFP TFs are shown to be potent regulators of gene expression with therapeutic promise in the treatment of disease.


Asunto(s)
Glioblastoma/metabolismo , Glioblastoma/terapia , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Dedos de Zinc/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/irrigación sanguínea , Glioblastoma/genética , Humanos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Transfección , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
9.
Oncogene ; 19(32): 3563-9, 2000 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-10951561

RESUMEN

The v-erbA oncoprotein (P75gag-v-erbA) can repress thyroid hormone receptor induced transcriptional activation of target genes. A central question is how hormone responsive elements in a target gene determine the transcriptional regulation mediated by P75gag-v-erbA. We addressed this with receptors chimeric between P75gag-v-erbA and thyroid hormone receptor (TR) by testing their regulatory activities on thyroid hormone response elements (TREs) differing in the sequence of the consensus core recognition motif AGGTCA. We report here that enhances, TR dependent transcriptional activation is conferred by P75gag-v-erbA when the thymidine in the half site recognition motif is exchanged for an adenosine. The enhancement was independent of the DNA binding region of P75gag-v-erbA, whereas increased expression of corepressor abolished the enhancing effect. The data indicate that the enhancement results from an impaired DNA binding by the oncoprotein combined with an effective scavenging of corepressors. Our data thus suggest the P75gag-v-erbA indirectly can contribute to enhancement of thyroid hormone induced gene expression.


Asunto(s)
Proteínas Oncogénicas v-erbA/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Elementos de Respuesta , Hormonas Tiroideas/metabolismo , Activación Transcripcional , Aminoácidos , Animales , Sitios de Unión , Línea Celular , ADN/metabolismo , Mutagénesis Sitio-Dirigida , Nucleótidos , Proteínas Oncogénicas v-erbA/genética , Codorniz , Receptores de Hormona Tiroidea/genética
10.
Oncogene ; 20(7): 775-87, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11314012

RESUMEN

v-ErbA is a mutated variant of thyroid hormone receptor (TRalpha/NR1A1) borne by the Avian Erythroblastosis virus causing erythroleukemia. TRalpha is known to activate transcription of specific genes in the presence of its cognate ligand, T3 hormone, while in its absence it represses it. v-ErbA is unable to bind ligand, and hence is thought to contribute to leukemogenesis by actively repressing erythroid-specific genes such as the carbonic anhydrase II gene (CA II). In the prevailing model, v-ErbA occludes liganded TR from binding to its cognate elements and constitutively interacts with the corepressors NCoR/SMRT. We previously identified a v-ErbA responsive element (VRE) within a DNase I hypersensitive region (HS2) located in the second intron of the CA II gene. We now show that HS2 fulfils all the requirements for a genuine enhancer that functions independent of its orientation and position with a profound erythroid-specific activity in normal erythroid progenitors (T2ECs) and in leukemic erythroid cell lines. We find that the HS2 enhancer activity is governed by two adjacent GATA-factor binding sites. v-ErbA as well as unliganded TR prevent HS2 activity by nullifying the positive function of factors bound to GATA-sites. However, v-ErbA, in contrast to TR, does not convey active repression to silence the transcriptional activity intrinsic to a heterologous tk promoter. We propose that depending on the sequence and context of the binding site, v-ErbA contributes to leukemogenesis by occluding liganded TR as well as unliganded TR thereby preventing activation or repression, respectively.


Asunto(s)
Anhidrasas Carbónicas/genética , Elementos de Facilitación Genéticos , Leucemia Eritroblástica Aguda/genética , Proteínas Oncogénicas v-erbA/genética , Receptores de Hormona Tiroidea/genética , Animales , Secuencia de Bases , Sitios de Unión , Pollos , Células Precursoras Eritroides , Regulación Leucémica de la Expresión Génica , Humanos , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Unión Proteica , Células Tumorales Cultivadas
11.
Oncogene ; 21(18): 2864-72, 2002 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-11973646

RESUMEN

T2EC are chicken erythrocytic progenitors that balance between self-renewal and differentiation as a function of response to specific growth factors. Their transformation by the v-erbA oncogene locks them into the self-renewal program. We show here that the expression of the VLA-2 integrin alpha2 subunit mRNA is downregulated by v-erbA and that VLA-2 engagement and clustering, brought about by treatment with an alpha2-specific antibody or by culture on the VLA-2 ligand collagen I, inhibits T2EC proliferation. From competition studies using antibodies, VLA-2 was shown to be involved in the collagen-induced response. While engagement of VLA-2 inhibited proliferation, it was not sufficient to induce differentiation. The transformation of T2EC by v-erbA decreased their interaction with collagen I and the VLA-2 brake on cell proliferation, which may account for the increased proliferation potential of transformed erythrocytic progenitors and for their shedding into the blood of infected chickens. Our data suggest that the interaction between erythroid progenitors and collagen, mediated by VLA-2, play a major role in the control of erythropoiesis in vitro and that this pathway is a target of the v-erbA oncogene.


Asunto(s)
Regulación hacia Abajo , Integrinas/genética , Proteínas Oncogénicas v-erbA/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Cationes Bivalentes , Adhesión Celular/fisiología , División Celular , Línea Celular , Pollos , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Eritrocitos/citología , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Células Madre Hematopoyéticas/citología , Integrinas/biosíntesis , Proteínas Oncogénicas v-erbA/genética , Receptores de Colágeno
12.
Oncogene ; 15(6): 701-15, 1997 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-9264411

RESUMEN

V-ErbA, a mutated thyroid hormone receptor (TR) alpha cooperates with tyrosine kinase oncoproteins to induce fatal erythroleukemia in chicks. In vitro, v-ErbA employs a similar cooperation to induce sustained proliferation and arrest differentiation of committed erythroid progenitors. V-ErbA has been proposed to function as a dominant-negative c-ErbA/TR alpha, since it lacks an AF-2 transactivation domain and cannot be activated by hormone but retains the capacity to bind corepressors. However, v-ErbA fails to heterodimerize with the coreceptor RXR, exhibits an altered DNA binding specificity and fails to suppress the action of coexpressed TR alpha/c-ErbA in erythroblasts. In this paper, we identify a novel mechanism by which v-ErbA contributes to leukemogenesis. Recently, the glucocorticoid receptor (GR) was identified as a key regulator of proliferation and differentiation in normal erythroid progenitors. For this, the GR required to cooperate with endogenous receptor tyrosine kinases (c-Kit) and with the estrogen receptor (ER). Here, we demonstrate that v-ErbA can substitute for the ligand-activated GR and ER, inducing proliferation and arresting differentiation in the presence of specific GR and ER antagonists. Like the GR, v-ErbA required to cooperate with c-Kit for both proliferation induction and differentiation arrest, being devoid of biological activity in the absence of an active c-Kit. In self-renewing erythroblasts, v-ErbA not only repressed known v-ErbA target genes but also maintained high expression of c-myb. These biological activities of v-ErbA depended on distinct mutations in the DNA-binding domain. Additionally, v-ErbA acted as a partial, weak repressor of c-ErbA/TR alpha function in normal erythroblasts. It could be converted into a truly dominant-negative receptor by restoring its ability to heterodimerize with RXR.


Asunto(s)
Proteínas Oncogénicas v-erbA/fisiología , Transformación Genética , Animales , Diferenciación Celular/genética , División Celular/genética , Células Cultivadas , Embrión de Pollo , Pollos , Eritroblastos , Fibroblastos , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos/genética , Cinética , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , Fosfotirosina/análisis , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-kit/fisiología , Proteínas Proto-Oncogénicas c-myb , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/fisiología , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/fisiología , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Receptores de Hormona Tiroidea/fisiología , Retroviridae/genética , Transactivadores/genética , Transactivadores/metabolismo
13.
Mol Endocrinol ; 11(3): 379-89, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9058383

RESUMEN

Several members of the thyroid hormone receptor (TR) family are able to switch from a transcriptional repressor to a transcriptional activator upon binding of their ligand. The oncogene v-erbA is a variant form of the TR unable to bind hormone and thus acts as a constitutive repressor. We demonstrate, using fusion proteins between the DNA-binding domain of the yeast factor GAL4 and the silencing domains of v-erbA and TR beta, that point mutations in three different regions severely affect their repression function. Furthermore, the three regions, each as an inactive fusion protein with the GAL4 DNA-binding domain, restore silencing activity when assembled on the same promoter. These observations define at least three silencing subdomains, SSD1-SSD3, which are involved in the silencing function of v-erbA. We propose a model in which full silencing activity is brought about by the combined interaction of each silencing subdomain with corepressors and/or basal transcription factors.


Asunto(s)
Mutagénesis Sitio-Dirigida , Proteínas Oncogénicas v-erbA/genética , Secuencia de Aminoácidos , Unión Competitiva , Regulación de la Expresión Génica , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Receptores de Hormona Tiroidea , Proteínas Recombinantes de Fusión/genética , Proteínas Represoras , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Factores de Transcripción , Transfección
14.
Mol Endocrinol ; 8(7): 819-28, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7984144

RESUMEN

The viral erb A oncogene is a mutated allele of a normal cell gene for a thyroid hormone receptor. The DNA recognition properties of the v-erb A protein are altered from those of the thyroid hormone receptor, due in part to a point mutation in the P-box of the zinc-finger domain of the viral allele. We report here the effects of systematically varying this P-box codon; our results suggest that this P-box amino acid contributes to DNA specificity not by promoting recognition of the appropriate response elements, but rather by excluding binding of the erb A protein to inappropriate half-sites. In this manner, DNA recognition by the v-erb A protein appears to differ from that by the glucocorticoid receptor. A variety of P-box amino acids were compatible with recognition of the prototypic AGGTCA half-site; intriguingly, several of these mutant erb A proteins could also recognize a variety of alternative half-site sequences. Recognition of these alternative half-sites required a compatible amino acid sequence in the N terminus of the erb A protein. Our results begin to define a code by which the identity of the amino acids in the zinc-finger and N-terminal domains is reflected in the DNA recognition properties of the receptor.


Asunto(s)
ADN/metabolismo , Proteínas Oncogénicas v-erbA/genética , Oncogenes , Mutación Puntual , Receptores de Hormona Tiroidea/genética , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Codón/genética , Secuencia de Consenso , ADN/genética , Regulación de la Expresión Génica , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Dedos de Zinc
15.
Mol Endocrinol ; 14(2): 201-11, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10674394

RESUMEN

Members of the thyroid hormone receptor (TR) family act on vertebrate development and homeostasis by activating or repressing transcription of specific target genes in a ligand-dependent way. Repression by TR in the absence of ligand is mediated by an active silencing mechanism. The oncogene v-ErbA is a variant form of TR unable to bind hormone and thus acts as a constitutive repressor. Functional studies and mutation analysis revealed that the TR/v-ErbA silencing domain is composed of three silencing subdomains (SSD1-3) which, although nonfunctional individually, synergize such that silencing activity is restored when they are combined in a heteromeric complex. Here we demonstrate, using protein interaction assays in vitro and in vivo, that the inactive v-ErbA point mutant L489R within helix 5/6 in SSD2 fails to interact with the two corepressors N-CoR (nuclear receptor corepressor) or SMRT (silencing mediator of retinoic acid and thyroid hormone receptor). Furthermore, mutants in SSD1 and SSD3 exhibit a reduced corepressor recruitment corresponding to their weak residual silencing activity. In mammalian two-hybrid assays, only the combination of all three silencing subdomains, SSD1-3, leads to a cooperative binding to the corepressors N-CoR or SMRT comparable to that of the full-length v-ErbA repression domain. In conclusion, full silencing activity requires corepressor interaction with all three silencing subdomains, SSD1-3. Among these, SSD2 is a new target for N-CoR and SMRT and is essential for corepressor binding and function.


Asunto(s)
Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Silenciador del Gen , Prueba de Complementación Genética , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear , Co-Represor 2 de Receptor Nuclear , Proteínas Oncogénicas v-erbA/química , Mutación Puntual , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética
16.
Mol Endocrinol ; 12(5): 645-53, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9605927

RESUMEN

The oncogenic counterpart of thyroid hormone receptor-alpha (TRRalpha), denoted P75gag-v-erbA, has served as a paradigm for the ability of TRs to repress basal levels of transcription. We show here that the retinoid X receptor (RXR), when activated by its specific ligand SR11237, is repressed by both the normal TRalpha and the P75gag-v-erbA. The repression caused by the two proteins is distinct and dependent on both the cell type and the hormone-response element through which RXR acts. In HeLa cells only TR repressed efficiently through the palindromic 2xIR0 element, whereas the proteins were equally efficient in JEG cells. This demonstrates that proteins distinct in the two cell types mediate the repression. RXR-dependent induction via the natural response element of the cellular retinol-binding protein (CRBPII) gene was likewise (> or = 50%) repressed by TR, whereas P75gag-v-erbA did not repress during the same conditions. Furthermore, P75gag-v-erbA and its variants v-erbAtd359 (lacking repressing activity on TR) and v-erbAr12 (a highly active repressor of TR) efficiently repressed induction by a hybrid protein consisting of the DNA- binding domain of Gal4 and the ligand-binding region of RXR. The viral proteins did not, however, associate with RXR unless the two partners were allowed to heterodimerize upon binding to a specific response element, such as the 2xIR0 element or that of the CRBPII gene. In conclusion, we suggest that the efficient repression seen with the the 2xIR0 element is due to heterodimerization of TR or the viral oncoproteins with RXR and a concomitant inhibition of binding of the RXR-specific ligand that results in an inability of RXR to attract a cell type-specific cofactor. In addition, the data suggest that the interaction between RXR and P75gag-v-erbA on the CRBPII element is too weak to inhibit RXR from binding a ligand and therefore also to repress.


Asunto(s)
Proteínas Oncogénicas v-erbA/fisiología , Proteínas Oncogénicas v-fos/fisiología , Receptores de Ácido Retinoico/fisiología , Receptores de Hormona Tiroidea/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Animales , Sitios de Unión/genética , Coriocarcinoma , ADN/metabolismo , Dimerización , Regulación de la Expresión Génica , Humanos , Ratones , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-fos/genética , Unión Proteica/genética , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Hormona Tiroidea/genética , Proteínas Represoras/genética , Receptores X Retinoide , Factores de Transcripción/antagonistas & inhibidores , Células Tumorales Cultivadas
17.
Mol Endocrinol ; 12(8): 1161-71, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9717842

RESUMEN

Nuclear hormone receptors are hormone-regulated transcription factors that bind to specific sites on DNA and modulate the expression of adjacent target genes. Many nuclear hormone receptors display bimodal transcriptional properties; thyroid hormone receptors, for example, typically repress target gene expression in the absence of hormone, but activate target gene expression in the presence of hormone. The ability to repress is closely linked to the ability of the apo-receptor to physically bind to auxiliary corepressor proteins denoted SMRT (silencing mediator of retinoic acid and thyroid hormone receptor) and N-CoR (nuclear receptor corepressor), which, in turn, help mediate the actual molecular events involved in transcriptional silencing. We report here that repression by thyroid hormone receptors can be regulated not only by cognate hormone, but also by certain tyrosine kinase signal transduction pathways, such as that represented by the epidermal growth factor-receptor. Activation of tyrosine kinase signaling leads to inhibition of T3R-mediated repression with relatively little effect on activation. These effects appear to be mediated by a kinase-initiated disruption of the ability of T3R to interact with SMRT corepressor. Intriguingly, tyrosine kinase signaling similarly disrupted the interactions of SMRT with v-Erb A, with retinoic acid receptors, and with PLZF, a nonreceptor transcriptional repressor. We conclude that tyrosine kinase signaling exerts potentially important regulatory effects on transcriptional silencing mediated by a variety of transcription factors that operate through the SMRT corepressor complex.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Receptores ErbB/metabolismo , Riñón/citología , Co-Represor 2 de Receptor Nuclear , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , Proteínas Oncogénicas v-erbB/genética , Proteínas Oncogénicas v-erbB/metabolismo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Transcripción Genética , Transfección
18.
J Mol Endocrinol ; 33(3): 585-608, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15591021

RESUMEN

Rev-erbalpha is a ubiquitously expressed orphan nuclear receptor which functions as a constitutive transcriptional repressor and is expressed in vertebrates according to a robust circadian rhythm. We report here that two Rev-erbalpha mRNA isoforms, namely Rev-erbalpha1 and Rev-erbalpha 2, are generated through alternative promoter usage and that both show a circadian expression pattern in an in vitro system using serum-shocked fibroblasts. Both promoter regions P1 (Rev-erbalpha1) and P2 (Rev-erbalpha2) contain several E-box DNA sequences which function as response elements for the core circadian-clock components: CLOCK and BMAL1. The CLOCK-BMAL1 heterodimer stimulates the activity of both P1 and P2 promoters in transient transfection assay by 3-6-fold. This activation was inhibited by the overexpression of CRY1, a component of the negative limb of the circadian transcriptional loop. Critical E-box elements were mapped within both promoters. This regulation is conserved in vertebrates since we found that the CLOCK-BMAL1 heterodimer also regulates the zebrafish Rev-erbalpha gene. In line with these data Rev-erbalpha circadian expression was strongly impaired in the livers of Clock mutant mice and in the pineal glands of zebrafish embryos treated with Clock and Bmal1 antisense oligonucleotides. Together these data demonstrate that CLOCK is a critical regulator of Rev-erbalpha circadian gene expression in evolutionarily distant vertebrates and suggest a role for Rev-erbalpha in the circadian clock output.


Asunto(s)
Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Proteínas Oncogénicas v-erbA/genética , Factores de Transcripción ARNTL , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Relojes Biológicos/genética , Proteínas CLOCK , Línea Celular , Ritmo Circadiano/genética , Dimerización , Humanos , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Elementos de Respuesta/genética , Alineación de Secuencia , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Pez Cebra/genética
19.
Mol Cell Endocrinol ; 199(1-2): 61-72, 2003 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-12581880

RESUMEN

Thyroid hormone receptor alpha (TRalpha) and the oncoprotein v-erbA (a mutated form of TRalpha incapable of binding T3) bind as heterodimers with retinoid X receptor (RXR) to DNA sequences with different orientations of AGGTCA half sites. v-erbA can also form homodimers, whereas, TRalpha1 homodimerizes poorly. Therefore, in order to obtain a better understanding for the distinct homodimerization properties between TRalpha1 and v-erbA, we created chimeras between these two receptors and tested their abilities to homodimerize on direct and everted repeats (DRs, ERs). We found that the enhanced homodimerization properties of v-erbA compared to TRalpha1 map to isoleucine at position 339 in conjunction with serine at position 351 and alanine at position 358. Our data indicate that the methyl group in isoleucine at position 339 plays an important role in v-erbA homodimerization, particularly on ER 6. Functional studies with I339V+S351P+A358T, a v-erbA mutant unable to homodimerize but still able to heterodimerize with RXR on ERs and DRs, indicate that v-erbA-RXR heterodimers mediate the dominant negative activity of v-erbA on DRs. However, the repressor activity of this mutant is weaker than that of the wild type v-erbA on ERs, suggesting that v-erbA homodimers rather than v-erbA-RXR heterodimers mediate the potent dominant negative activity of v-erbA on ERs.


Asunto(s)
Proteínas Oncogénicas v-erbA/genética , Receptores alfa de Hormona Tiroidea/genética , Secuencia de Aminoácidos/fisiología , Animales , Dimerización , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Oncogénicas v-erbA/química , Proteínas Oncogénicas v-erbA/metabolismo , Mutación Puntual , Estructura Secundaria de Proteína , Receptores de Ácido Retinoico/metabolismo , Proteínas Recombinantes de Fusión/genética , Receptores X Retinoide , Alineación de Secuencia , Receptores alfa de Hormona Tiroidea/química , Receptores alfa de Hormona Tiroidea/metabolismo , Factores de Transcripción/metabolismo , Transfección
20.
J Steroid Biochem Mol Biol ; 67(3): 181-91, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9879977

RESUMEN

The v-erbA oncogene protein, p75(gag-v-erbA), is a mutant form of the thyroid hormone receptor alpha (TR alpha) which has sustained mutations both in the ligand binding and DNA binding domains. The oncoprotein has therefore lost its ability to bind ligand, and its heterodimerization with the retinoid-X receptor (RXR) is impaired. Here, we have investigated the effects of the mutations in the DNA binding domain. By applying a PCR-based screening assay we isolated DNA sequences to which p75(gag-v-erbA) binds as a heterodimer with RXR, and characterized these with regard to their nucleotide sequence and ability to associate with RXR/P75(gag-v-erbA) heterodimers in vitro and in vivo. In the PCR selection assay the heterodimer exhibited a preference for direct repeats with a 3' half-site sequence AGGTCG and spacers of four or five nucleotides separating the two half-sites. These DNA binding data were confirmed by gel retardation assays with synthetic oligonucleotides as well as by transfection experiments using dominantly active VP16 fusion proteins with P75(gag-v-erbA) and TR alpha. The comparison between RXR/P75(gag-v-erbA) and RXR/TR alpha heterodimers demonstrated that although their DNA binding properties are very similar, however, a relaxed specificity of P75(gag-v-erbA) for the spacer length may allow it to interfere with more hormone signalling pathways than only that of thyroid hormone.


Asunto(s)
ADN/genética , ADN/metabolismo , Proteínas Oncogénicas v-erbA/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , Cartilla de ADN/genética , Dimerización , Células HeLa , Humanos , Técnicas In Vitro , Mutación , Proteínas Oncogénicas v-erbA/química , Proteínas Oncogénicas v-erbA/genética , Reacción en Cadena de la Polimerasa , Receptores de Ácido Retinoico/química , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptores de Hormona Tiroidea/química , Receptores de Hormona Tiroidea/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Receptores X Retinoide , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Virus Vaccinia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA