Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Endocrinol ; 539: 111415, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34339825

RESUMEN

Sex hormones, such as androgens, estrogens and progestins are naturally occurring compounds that tightly regulate endocrine systems in a variety of living organisms. Uncontrolled environmental exposure to these hormones or their biological and synthetic mimetics has been widely documented. Furthermore, water contaminants penetrate soil to affect flora, fauna and ultimately humans. Because endocrine systems evolved to respond to very small changes in hormone levels, the low levels found in the environment cannot be ignored. The combined actions of sex hormones with glucocorticoids and other nuclear receptors disruptors creates additional level of complexity including the newly described "dynamic assisted loading" mechanism. We reviewed the extensive literature pertaining to world-wide detection of these disruptors and created a detailed Table on the development and current status of methods used for their analysis.


Asunto(s)
Disruptores Endocrinos/efectos adversos , Hormonas Esteroides Gonadales/efectos adversos , Animales , Disruptores Endocrinos/análisis , Glucocorticoides/efectos adversos , Humanos , Contaminantes Químicos del Agua/efectos adversos , Contaminantes Químicos del Agua/análisis
2.
Br J Cancer ; 105(10): 1593-9, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22009029

RESUMEN

BACKGROUND: We aimed to evaluate the clinical relevance of p53 and p73 isoforms that modulate the function of p53. METHODS: This prospective multicentre study included 154 patients with stage III and IV serous ovarian cancer. A functional yeast-based assay and subsequent sequencing were performed to analyse the p53 mutational status. Expression of p53 and p73 isoforms was determined using RT-qPCR. RESULTS: Δ133p53 expression constituted an independent prognostic marker for recurrence-free (hazard ratio=0.571, P=0.016, 95% CI: 0.362-0.899) and overall survival (hazard ratio=0.365, P=0.004, 95% CI: 0.182-0.731) in patients with p53 mutant ovarian cancer (n=121). High Δ40p53 expression was associated with favourable tumour grading (P=0.037) and improved recurrence-free survival (33.4 vs 19.6 months, P=0.029), but not overall survival (43.1 vs 33.6 months, P=0.139), in patients with p53 wild-type cancer (n=33). Neither the p53 mutational status nor p73 isoform expression possessed prognostic significance in the examined ovarian cancer cases. CONCLUSION: Δ133p53 expression was associated with prognosis in the vast majority of ovarian cancer cases, that is, patients with p53 mutant advanced serous carcinomas. Thus, our findings underline the importance of considering the complex p53 regulatory network.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Ováricas/patología , Pronóstico , Proteína p53 Supresora de Tumor/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Genes p53 , Humanos , Persona de Mediana Edad , Mutación , Neoplasias Ováricas/metabolismo , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína p53 Supresora de Tumor/genética
3.
Pharmaceuticals (Basel) ; 14(4)2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33917884

RESUMEN

Inflammasome targeting and controlling dysbiosis are promising therapeutic approaches to control ulcerative colitis. This report is the first to investigate the mechanisms underlying the coloprotective effects of rosuvastatin and Lactobacillus and their combined therapy on dextran sodium sulfate (DSS)-induced colitis in high-fat diet (HFD)-fed rats. Our results demonstrate the aggravation of intestinal inflammation as a consequence of an HFD following DSS administration. An association between dyslipidemia, LDL oxidation, CD36 expression, ROS generation, thioredoxin-interacting protein (TXNIP) upregulation, and NLRP3 inflammasome activation was demonstrated by DSS exposure in HFD-fed rats. We demonstrated that rosuvastatin/Lactobacillus significantly suppressed the DSS/HFD-induced increase in colon weight/length ratio, DAI, MDI, and myeloperoxidase, as well as corrected dysbiosis and improved histological characteristics. Additionally, caspase-1 activity and IL-1ß-driven pyroptotic activity was significantly reduced. Rosuvastatin/Lactobacillus showed prominent anti-inflammatory effects as revealed by the IL-10/IL-12 ratio and the levels of TNF-α and IL-6. These latter effects may be attributed to the inhibition of phosphorylation-induced activation of NF-κB and a concomitant reduction in the expression of NLRP3, pro-IL-1ß, and pro-IL-18. Furthermore, rosuvastatin/Lactobacillus reduced Ox-LDL-induced TXNIP and attenuated the inflammatory response by inhibiting NLRP3 inflammasome assembly. To conclude, rosuvastatin/Lactobacillus offers a safe and effective strategy for the management of ulcerative colitis.

5.
J Cell Biol ; 154(1): 33-48, 2001 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-11448988

RESUMEN

We have examined the relationship between transcription and chromatin structure using a tandem array of the mouse mammary tumor virus (MMTV) promoter driving a ras reporter. The array was visualized as a distinctive fluorescent structure in live cells stably transformed with a green fluorescent protein (GFP)-tagged glucocorticoid receptor (GR), which localizes to the repeated MMTV elements after steroid hormone treatment. Also found at the array by immunofluorescence were two different steroid receptor coactivators (SRC1 and CBP) with acetyltransferase activity, a chromatin remodeler (BRG1), and two transcription factors (NFI and AP-2). Within 3 h after hormone addition, arrays visualized by GFP-GR or DNA fluorescent in situ hybridization (FISH) decondensed to varying degrees, in the most pronounced cases from a approximately 0.5-microm spot to form a fiber 1-10 microm long. Arrays later recondensed by 3-8 h of hormone treatment. The degree of decondensation was proportional to the amount of transcript produced by the array as detected by RNA FISH. Decondensation was blocked by two different drugs that inhibit polymerase II, 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) and alpha-amanitin. These observations demonstrate a role for polymerase in producing and maintaining decondensed chromatin. They also support fiber-packing models of higher order structure and suggest that transcription from a natural promoter may occur at much higher DNA-packing densities than reported previously.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética , Amanitinas/farmacología , Animales , Proteínas Portadoras/metabolismo , Cromatina/ultraestructura , ADN/metabolismo , ADN Helicasas , Proteínas de Unión al ADN/metabolismo , Diclororribofuranosil Benzoimidazol/farmacología , Inhibidores Enzimáticos/farmacología , Genes ras/genética , Proteínas Fluorescentes Verdes , Histona Acetiltransferasas , Hibridación Fluorescente in Situ , Proteínas Luminiscentes/metabolismo , Virus del Tumor Mamario del Ratón/genética , Ratones , Microscopía Fluorescente , Factores de Transcripción NFI , Proteínas Nucleares/metabolismo , Coactivador 1 de Receptor Nuclear , Receptores de Glucocorticoides/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Factor de Transcripción AP-2 , Factores de Transcripción/metabolismo , Transfección
6.
Science ; 255(5051): 1573-6, 1992 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-1347958

RESUMEN

The mouse mammary tumor virus (MMTV) promoter attains a phased array of six nucleosomes when introduced into rodent cells. This architecture excludes nuclear factor 1/CCAAT transcription factor (NF1/CTF) from the promoter before glucocorticoid treatment and hormone-dependent access of nucleolytic agents to promoter DNA. In contrast, when the promoter was transiently introduced into cells, NF1/CTF was bound constitutively and nucleolytic attack was hormone-independent. Thus, induction at this promoter was a bimodal process involving receptor-dependent remodeling of chromatin that allows NF1/CTF loading and direct receptor-mediated recruitment of additional transcription factors.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Regulación Viral de la Expresión Génica , Virus del Tumor Mamario del Ratón/genética , Regiones Promotoras Genéticas/fisiología , Roedores/genética , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , ADN Viral/fisiología , Proteínas de Unión al ADN/fisiología , Dexametasona/farmacología , Glucocorticoides/fisiología , Técnicas In Vitro , Datos de Secuencia Molecular , Factores de Transcripción NFI , Proteínas Nucleares , Nucleosomas/fisiología , Polimorfismo de Longitud del Fragmento de Restricción , Mapeo Restrictivo , Transfección , Proteína 1 de Unión a la Caja Y
7.
Science ; 287(5456): 1262-5, 2000 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-10678832

RESUMEN

Steroid receptors bind to site-specific response elements in chromatin and modulate gene expression in a hormone-dependent fashion. With the use of a tandem array of mouse mammary tumor virus reporter elements and a form of glucocorticoid receptor labeled with green fluorescent protein, targeting of the receptor to response elements in live mouse cells was observed. Photobleaching experiments provide direct evidence that the hormone-occupied receptor undergoes rapid exchange between chromatin and the nucleoplasmic compartment. Thus, the interaction of regulatory proteins with target sites in chromatin is a more dynamic process than previously believed.


Asunto(s)
Cromatina/metabolismo , Dexametasona/farmacología , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta , Secuencias Repetidas Terminales , Animales , Sitios de Unión , Línea Celular Transformada , Núcleo Celular/metabolismo , Dexametasona/metabolismo , Proteínas Fluorescentes Verdes , Hibridación Fluorescente in Situ , Ligandos , Proteínas Luminiscentes , Virus del Tumor Mamario del Ratón/genética , Ratones , Microscopía Confocal , Microscopía Fluorescente , Nucleosomas/metabolismo
8.
Curr Biol ; 11(24): 1981-5, 2001 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-11747826

RESUMEN

Activation of gene transcription involves chromatin remodeling by coactivator proteins that are recruited by DNA-bound transcription factors. Local modification of chromatin structure at specific gene promoters by ATP-dependent processes and by posttranslational modifications of histone N-terminal tails provides access to RNA polymerase II and its accompanying transcription initiation complex. While the roles of lysine acetylation, serine phosphorylation, and lysine methylation of histones in chromatin remodeling are beginning to emerge, low levels of arginine methylation of histones have only recently been documented, and its physiological role is unknown. The coactivator CARM1 methylates histone H3 at Arg17 and Arg26 in vitro and cooperates synergistically with p160-type coactivators (e.g., GRIP1, SRC-1, ACTR) and coactivators with histone acetyltransferase activity (e.g., p300, CBP) to enhance gene activation by steroid and nuclear hormone receptors (NR) in transient transfection assays. In the current study, CARM1 cooperated with GRIP1 to enhance steroid hormone-dependent activation of stably integrated mouse mammary tumor virus (MMTV) promoters, and this coactivator function required the methyltransferase activity of CARM1. Chromatin immunoprecipitation assays and immunofluorescence studies indicated that CARM1 and the CARM1-methylated form of histone H3 specifically associated with a large tandem array of MMTV promoters in a hormone-dependent manner. Thus, arginine-specific histone methylation by CARM1 is an important part of the transcriptional activation process.


Asunto(s)
Arginina/metabolismo , Histonas/metabolismo , Hormonas/fisiología , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/fisiología , Esteroides/fisiología , Acetilación , Técnica del Anticuerpo Fluorescente , Histonas/química , Lisina/metabolismo , Virus del Tumor Mamario del Ratón/genética , Metilación , Fosforilación , Pruebas de Precipitina , Serina/metabolismo
9.
Mol Cell Biol ; 15(4): 2125-34, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7891707

RESUMEN

Recent studies have provided evidence of crosstalk between steroid receptors and cyclic AMP (cAMP) signalling pathways in the regulation of gene expression. A synergism between intracellular phosphorylation inducers and either glucocorticoids or progestins has been shown to occur during activation of the mouse mammary tumor virus (MMTV) promoter. We have investigated the effect of 8-Br-cAMP and okadaic acid, modulators of cellular kinases and phosphatases, on the hormone-induced activation of the MMTV promoter in two forms: a transiently transfected template with a disorganized, accessible nucleoprotein structure and a stably replicating template with an ordered, inaccessible nucleoprotein structure. Both okadaic acid and 8-Br-cAMP synergize significantly with either glucocorticoids or progestins in activating the transiently transfected MMTV template. In contrast, 8-Br-cAMP, but not okadaic acid, is antagonistic to hormone-induced activation of the stably replicating MMTV template. Nuclear run-on experiments demonstrate that this inhibition is a transcriptional effect on both hormone-induced transcription and basal transcription. Surprisingly, 8-Br-cAMP does not inhibit glucocorticoid-induced changes in restriction enzyme access and nuclear factor 1 binding. However, association of a complex with the TATA box region is inhibited in the presence of 8-Br-cAMP. Thus, cAMP treatment interferes with the initiation process but does not inhibit interaction of the receptor with the template. Since the replicated, ordered MMTV templates and the transfected, disorganized templates show opposite responses to 8-Br-cAMP treatment, we conclude that chromatin structure can influence the response of a promoter to activation of the cAMP signalling pathway.


Asunto(s)
Virus del Tumor Mamario del Ratón/genética , Nucleoproteínas/metabolismo , Regiones Promotoras Genéticas/genética , Transducción de Señal/genética , Transcripción Genética , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Dexametasona/farmacología , Éteres Cíclicos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Modelos Genéticos , Ácido Ocadaico , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
10.
Mol Cell Biol ; 9(7): 3127-31, 1989 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2550796

RESUMEN

Specific DNA sequence elements which contain binding sites for the glucocorticoid receptor mediate the action of glucocorticoid hormones on gene transcription. In glucocorticoid-inducible genes, these glucocorticoid-responsive elements behave as hormone-inducible enhancers of transcription. We have taken advantage of the bovine papillomavirus (BPV) system to test the stringency of glucocorticoid regulation of transcription. BPV episomes were constructed to contain two hormone-regulated transcription units in close proximity; one transcription unit is under control of a glucocorticoid-inducible promoter (mouse mammary tumor virus) while the other is under control of a glucocorticoid-inhibited promoter (pro-opiomelanocortin). Glucocorticoids independently regulated transcription of the two physically linked transcription units, irrespective of their relative orientation and of their proximity on the BPV episomes. This result contrasts with the so-called position-independent activity of enhancers and suggests that the multicomponent organization of eucaryotic promoters restricts the action of hormone-responsive regulatory elements to a specific transcription unit, thus accounting for the stringency of hormonal regulation observed in vivo.


Asunto(s)
Glucocorticoides/fisiología , Virus del Tumor Mamario del Ratón/genética , Proopiomelanocortina/genética , Regiones Promotoras Genéticas , Animales , Línea Celular Transformada , ADN/genética , Immunoblotting , Papillomaviridae/genética , Plásmidos , Ratas
11.
Mol Cell Biol ; 11(2): 688-98, 1991 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1846670

RESUMEN

A fragment of the mouse mammary tumor virus (MMTV) promoter was reconstituted from pure histones into a dinucleosome with uniquely positioned octamer cores. Core boundaries for the in vitro-assembled dinucleosome corresponded to the observed in vivo phasing pattern for long terminal repeat nucleosomes A and B. Nuclear factor 1 (NF1), a constituent of the MMTV transcription initiation complex, was excluded from the assembled dinucleosome, whereas the glucocorticoid receptor was able to bind. During transcription of MMTV in vivo, displacement of nucleosome B was necessary to permit assembly of the initiation complex. These results indicate that the nucleoprotein structure of the promoter can provide differential access to sequence-specific DNA-binding proteins and that active chromatin remodeling can occur during transcription activation.


Asunto(s)
Virus del Tumor Mamario del Ratón/genética , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromatina/ultraestructura , ADN Viral/genética , Histonas/aislamiento & purificación , Histonas/metabolismo , Ratones , Conformación de Ácido Nucleico , Secuencias Repetitivas de Ácidos Nucleicos , Mapeo Restrictivo , Factor de Transcripción TFIID
12.
Mol Cell Biol ; 11(5): 2529-37, 1991 May.
Artículo en Inglés | MEDLINE | ID: mdl-1708094

RESUMEN

In vivo expression of the mouse mammary tumor virus (MMTV) is restricted to a few organs, with the highest rate of transcription found in the mammary gland. Using a series of mammary and nonmammary murine cell lines, we have identified two regulatory elements, located upstream of the hormone responsive element, that specifically regulate the MMTV promoter. The first element displays an enhancerlike activity and is coincident with the binding of a nuclear factor (designated MP4; position -1078 to -1052 in the long terminal repeat) whose presence is apparently restricted to mammary cell lines. The second regulatory region mediates a repressive activity and is mapped to the long terminal repeat segment from -415 to -483. This repression is specific for a particular subtype of mammary cells (RAC cells) able to grow under two differentiation states (A. Sonnenberg, H. Daams, J. Calafat, and J. Hilgers, Cancer Res. 46:5913-5922, 1986). The MMTV promoter in mammary cell lines thus appears to be modulated by two cis-acting elements that are likely to be involved in tissue-specific expression in vivo.


Asunto(s)
Regulación Viral de la Expresión Génica , Genes Virales , Virus del Tumor Mamario del Ratón/genética , Regiones Promotoras Genéticas , Secuencias Repetitivas de Ácidos Nucleicos , Animales , Secuencia de Bases , Línea Celular , Núcleo Celular/metabolismo , Deleción Cromosómica , Desoxirribonucleasa I , Dexametasona/farmacología , Ratones , Datos de Secuencia Molecular , Mapeo Nucleótido , Plásmidos , ARN/genética , ARN/aislamiento & purificación , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Mapeo Restrictivo , Transfección
13.
Mol Cell Biol ; 15(1): 26-34, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7799933

RESUMEN

We have stably introduced a reporter gene under the control of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR) into human T47D breast cancer cells to study the action of the progesterone receptor (PR) on transcription from a chromatin template. Unexpectedly, the chromatin organization of the MMTV LTR in these human breast cancer cells differed markedly from what we have observed previously. The region adjacent to the transcription start site (-221 to -75) was found to be constitutively hypersensitive to restriction enzyme cleavage in the absence of hormone. This region is normally encompassed within the second nucleosome of a phased array of six nucleosomes that is assembled when the MMTV LTR is stably maintained in mouse cells. Characteristically, in these rodent cells, the identical DNA sequences show increased restriction enzyme cleavage only in the presence of glucocorticoid. The increased access of restriction enzymes observed in the human PR+ cells was not observed in adjacent nucleosomes and was unaffected by treatment with the progesterone antagonist RU486. In addition, exonuclease III-dependent stops corresponding to the binding sites for nuclear factor 1 and the PR were observed before and after hormone treatment. These results indicate that MMTV chromatin replicated in these cells is organized into a constitutively open architecture and that this open chromatin state is accompanied by hormone-independent loading of a transcription factor complex that is normally excluded from uninduced chromatin.


Asunto(s)
Neoplasias de la Mama/genética , Cromatina/ultraestructura , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Virus del Tumor Mamario del Ratón/genética , Regiones Promotoras Genéticas , Humanos , Técnicas In Vitro , Nucleasa Microcócica/metabolismo , Nucleosomas/ultraestructura , Promegestona/farmacología , ARN Mensajero/genética , Secuencias Repetitivas de Ácidos Nucleicos , Mapeo Restrictivo , Transcripción Genética/efectos de los fármacos , Transfección , Células Tumorales Cultivadas
14.
Mol Cell Biol ; 18(6): 3633-44, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9584204

RESUMEN

Stimulation of the mouse mammary tumor virus with steroids results in the generation of a DNase I-hypersensitive region (HSR) spanning the hormone responsive element (HRE) in the long terminal repeat. Restriction enzymes were used to characterize the accessibility of various sites within the HSR of mouse mammary tumor virus long terminal repeat-reporter constructions in four different cell lines. The glucocorticoid-dependent HSR was found to span minimally 187 bases, a stretch of DNA longer than that associated with histones in the core particle. Although the 5'-most receptor binding site within the HRE is downstream of -190, hypersensitive sites were found further upstream to at least -295. The relationship in the accessibility between pairs of sites in the vicinity of the HSR was further examined in one cell line by a two-enzyme restriction access assay. In the uninduced state, the accessibilities at these sites were found to be independent of each other. In contrast, when stimulated with hormone, the accessibilities at these sites were observed to become linked. That is, once a distinct promoter was activated, all of the sites within the HSR of that molecule became accessible. The HSR formed along an invariant stretch of DNA sequence despite the multiplicity of nucleosome frames in the nucleosome B region, where the HRE is located. The results indicate that the macroscopic length of the HSR does not arise from core length-remodeling events in molecules containing Nuc-B in alternative positions.


Asunto(s)
Glucocorticoides/metabolismo , Virus del Tumor Mamario del Ratón/genética , Nucleosomas/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Animales , Línea Celular , ADN Viral/metabolismo , Desoxirribonucleasa I/metabolismo , Ligamiento Genético , Ratones , Conformación de Ácido Nucleico , Mapeo Restrictivo , Análisis de Secuencia de ADN , Relación Estructura-Actividad , Activación Transcripcional
15.
Mol Cell Biol ; 7(2): 830-7, 1987 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3102946

RESUMEN

Using three independent approaches, we studied the effects of H-ras on metastasis formation. Analysis of five in vitro-ras-transfected 10T1/2 clones with either flat or refractile morphologies revealed a relationship between metastatic potential, H-ras expression, and anchorage-independent growth. Four metastatic variants derived from a poorly metastatic, low-H-ras-expressing line all expressed high levels of H-ras RNA and grew efficiently in soft agar. Activation of H-ras expression in the metastatic tumors had occurred through amplification and rearrangement of H-ras sequences. In addition, preinduction of p21 synthesis in NIH 3T3 line 433, which contains v-H-ras under transcriptional control of the glucocorticoid-sensitive mouse mammary tumor virus long terminal repeat, significantly increased metastatic efficiency. Glucocorticoid treatment of normal or pEJ-transformed NIH 3T3 cells did not affect metastatic potential. These data reveal a direct relationship between ras expression and metastasis formation and suggest that metastatic and transformed phenotypes may be coregulated in ras-transformed 10T1/2 and NIH 3T3 cells.


Asunto(s)
Metástasis de la Neoplasia , Neoplasias Experimentales/patología , Oncogenes , Animales , Línea Celular , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos , Neoplasias Experimentales/genética , ARN Mensajero/genética
16.
Mol Cell Biol ; 17(11): 6481-90, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9343411

RESUMEN

Transcription of the retinoic acid receptor beta2 (RARbeta2) gene is induced by retinoic acid (RA) in mouse P19 embryonal carcinoma (EC) cells. Here we studied RA-induced chromatin structure alterations in the endogenous RARbeta2 promoter and in an integrated, multicopy RARbeta2 promoter in EC cells. RA markedly increased restriction site accessibility within the promoter, including a site near the RA responsive element (RARE) to which the nuclear receptor retinoid X receptor (RXR)-RAR heterodimer binds. These changes coincided with RA-induced alterations in the DNase I hypersensitivity pattern in and around the promoter. These changes became undetectable upon removal of RA, which coincided with the extinction of transcription. Analyses with receptor-selective ligands and an antagonist showed that increase in restriction site accessibility correlates with transcriptional activation, which parallels the RA-induced in vivo footprint of the promoter. Despite these changes, the micrococcal nuclease digestion profile of this promoter was not altered by RA. These results indicate that concurrent with the binding of the RXR-RAR heterodimer to the RARE, the local chromatin structure undergoes dynamic, reversible changes in and around the promoter without globally affecting the nucleosomal organization.


Asunto(s)
Cromatina/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Receptores de Ácido Retinoico/genética , Tretinoina/farmacología , Animales , Benzoatos/farmacología , Cromanos/farmacología , Huella de ADN , Regulación Neoplásica de la Expresión Génica , Ligandos , Ratones , Nucleasa Microcócica/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Transcripción Genética , Células Tumorales Cultivadas
17.
Mol Cell Biol ; 3(11): 2045-57, 1983 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-6318079

RESUMEN

The mouse mammary tumor virus long terminal repeat (MMTV LTR) has been introduced into cultured murine cells, using the 69% transforming fragment of bovine papilloma virus type 1 (BPV). Transformed cells contain up to 200 copies of the chimeric molecules per diploid genome. The restriction endonuclease map of the acquired recombinants, as well as the physical structure of the DNA, indicates that the LTR-BPV molecules present in these cells occur exclusively as unintegrated, extrachromosomal episome. When a 72-base pair direct repeat "enhancer" element (derived from the Harvey sarcoma retrovirus) was included in the MMTV LTR-BPV chimeric plasmids, DNA acquired through transfection, with a single exception, was integrated or rearranged or both. The transcriptional potential of the episomal MMTV promoter present in these cells was tested in two ways. First, steady-state levels of MMTV-initiated RNA were measured by quantitative S1 mapping. Second, the relative number of transcription complexes initiated in vivo was determined by using a subnuclear fraction highly enriched for MMTV-BPV minichromosomes in an in vitro transcription extension assay. Both approaches showed that the MMTV LTR present in the episomal state was capable of supporting glucocorticoid hormone-regulated transcription. We have therefore demonstrated the hormone response for the first time in a totally defined primary sequence environment. Significant differences both in the basal level of MMTV-initiated transcription and in the extent of glucocorticoid induction were observed in individual cell lines with similar episomal copy numbers. These phenotypic variations suggest that epigenetic structure is an important component of the mechanism of regulation.


Asunto(s)
Glucocorticoides/farmacología , Virus del Tumor Mamario del Ratón/genética , Operón , Transcripción Genética/efectos de los fármacos , Animales , Papillomavirus Bovino 1/genética , Transformación Celular Viral , Células Cultivadas , ADN Viral/genética , Amplificación de Genes , Ratones , Plásmidos , ARN Viral/genética , Secuencias Repetitivas de Ácidos Nucleicos , Transfección
18.
Mol Cell Biol ; 19(12): 8146-57, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10567540

RESUMEN

We utilized the mouse mammary tumor virus (MMTV) long terminal repeat (LTR) in vivo to understand how the interaction of the glucocorticoid receptor (GR) with a nucleosome-assembled promoter allows access of factors required for the transition from a repressed promoter to a derepressed, transcriptionally competent promoter. A mutation (C644G) in the ligand binding domain (LBD) of the mouse GR has provided information regarding the steps required in the derepression/activation process and in the functional significance of the two major transcriptional activation domains, AF1 and AF2. The mutant GR activates transcription from a transiently transfected promoter that has a disordered nucleosomal structure, though significantly less well than the wild-type GR. With an integrated, replicated promoter, which is assembled in an ordered nucleosomal array, the mutant GR does not activate transcription, and it fails to induce chromatin remodeling of the MMTV LTR promoter, as indicated by nuclease accessibility assays. Together, these findings support a two-step model for the transition of a nucleosome-assembled, repressed promoter to its transcriptionally active, derepressed form. In addition, we find that the C-terminal GR mutation is dominant over the transcription activation function of the N-terminal GR activation domain. These findings suggest that the primary activation function of the C-terminal activation domain is different from the function of the N-terminal activation domain and that it is required for derepression of the chromatin-repressed MMTV promoter.


Asunto(s)
Cromatina/fisiología , Receptores de Glucocorticoides/fisiología , Activación Transcripcional , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Genes Reporteros , Glucocorticoides/metabolismo , Ligandos , Virus del Tumor Mamario del Ratón/genética , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Ratas , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Endonucleasas Específicas del ADN y ARN con un Solo Filamento/metabolismo , Secuencias Repetidas Terminales , Triamcinolona Acetonida/metabolismo , Células Tumorales Cultivadas
19.
Mol Cell Biol ; 2(11): 1331-8, 1982 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-6298596

RESUMEN

NTRE 7 is an avian retrovirus recombinant of the endogenous nononcogenic Rous-associated virus-0 (RAV-0) and the oncogenic, exogenous, transformation-defective (td) Prague strain of Rous sarcoma virus B (td-PrRSV-B). Oligonucleotide mapping had shown that the recombinant virus is indistinguishable from its RAV-0 parent except for the 3'-end sequences, which were derived from td-PrRSV-B. However, the virus exhibits properties which are typical of an exogenous virus: it grows to high titers in tissue culture, and it is oncogenic in vivo. To accurately define the genetic region responsible for these properties, we determined the nucleotide sequences of the recombinant and its RAV-0 parent by using molecular clones of their DNA. These were compared with sequences already available for PrRSV-C, a virus closely related to the exogenous parent td-PrRSV-B. The results suggested that the crossover event which generated NTRE 7 took place in a region -501 to -401 nucleotides from the 3' end of the td-PrRSV parental genome and that sequences to the right of the recombination region were responsible for its growth properties and oncogenic potential. These sequences included a 148-base-pair exogenous-virus-specific region that was absent from the RAV-0 genome and the U3 region of the long terminal repeat. Since the exogenous-virus-specific sequences are expected to be missing from transformation-defective mutants of the Schmidt-Ruppin strain of RSV, which, like other exogenous viruses, grow to high titers in tissue culture and are oncogenic in vivo, we concluded that the growth properties and oncogenic potential of the exogenous viruses are determined by sequences in the U3 region of the long terminal repeat. However, we propose that the exogenous-virus-specific region may play a role in determining the oncogenic spectrum of a given oncogenic virus.


Asunto(s)
Virus del Sarcoma Aviar/genética , Transformación Celular Neoplásica , Genes Virales , Recombinación Genética , Secuencias Repetitivas de Ácidos Nucleicos , Animales , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/fisiología , Virus del Sarcoma Aviar/fisiología , Secuencia de Bases , Línea Celular , Transformación Celular Viral , Coturnix , Replicación Viral
20.
Mol Cell Biol ; 18(4): 2184-95, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9528790

RESUMEN

We have previously described a 160-bp enhancer (BCE-1) in the bovine beta-casein gene that is activated in the presence of prolactin and extracellular matrix (ECM). Here we report the characterization of the enhancer by deletion and site-directed mutagenesis, electrophoretic mobility shift analysis, and in vivo footprinting. Two essential regions were identified by analysis of mutant constructions: one binds C/EBP-beta and the other binds MGF/STAT5 and an as-yet-unidentified binding protein. However, no qualitative or quantitative differences in the binding of these proteins were observed in electrophoretic mobility shift analysis using nuclear extracts derived from cells cultured in the presence or absence of ECM with or without prolactin, indicating that prolactin- and ECM-induced transcription was not dependent on the availability of these factors in the functional cell lines employed. An in vivo footprinting analysis of the factors bound to nuclear chromatin in the presence or absence of ECM and/or prolactin found no differences in the binding of C/EBP-beta but did not provide definitive results for the other factors. Neither ECM nor prolactin activated BCE-1 in transient transfections, suggesting that the chromosomal structure of the integrated template may be required for ECM-induced transcription. Further evidence is that treatment of cells with inhibitors of histone deacetylase was sufficient to induce transcription of integrated BCE-1 in the absence of ECM. Together, these results suggest that the ECM induces a complex interaction between the enhancer-bound transcription factors, the basal transcriptional machinery, and a chromosomally integrated template responsive to the acetylation state of the histones.


Asunto(s)
Caseínas/genética , Elementos de Facilitación Genéticos , Matriz Extracelular/fisiología , Regulación de la Expresión Génica , Histonas/metabolismo , Proteínas de la Leche , Prolactina/fisiología , Acetilación , Animales , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT , Bovinos , Línea Celular , Cromatina/metabolismo , ADN , Proteínas de Unión al ADN/metabolismo , Electroforesis en Gel de Poliacrilamida , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/metabolismo , Unión Proteica , Factor de Transcripción STAT5 , Eliminación de Secuencia , Moldes Genéticos , Transactivadores/metabolismo , Activación Transcripcional , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA