Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
4.
Endocr J ; 58(2): 77-85, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21242649

RESUMEN

A human multi-protein complex (WINAC), composed of SWI/SNF components and DNA replication-related factors, that directly interacts with the vitamin D receptor (VDR) through the Williams syndrome transcription factor (WSTF), was identified with an ATP-dependent chromatin remodeling activity. This novel ATP-dependent chromatin remodeling complex facilitates VDR-mediated transrepression as well as transactivation with its ATP-dependent chromatin remodeling activity and promoter targeting property for the activator to access to the DNA. It also suggested that in this complex, WSTF serves as a signaling sensor to receive intra-cellular singalings to switch the activity of WINAC as well as WICH, another ATP-dependent chromatin remodeling complex containing hSNF2h. By making WSTF-deficient mice, some of the heart defects as well as abnormal calcium metabolism observed in Williams syndrome are attributed to the abnormal chromatin remodeling activity caused by WSTF deficiency. Thus, we would propose to designate Williams syndrome as an epigenome-regulator disease.


Asunto(s)
Ensamble y Desensamble de Cromatina , Síndrome de Williams/genética , Adenosina Trifosfato/farmacología , Animales , Anomalías Cardiovasculares , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Cromosomas Humanos Par 7/genética , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Trastornos del Crecimiento , Humanos , Hipercalcemia , Discapacidad Intelectual , Ratones , Ratones Noqueados , Receptores de Calcitriol/genética , Receptores de Calcitriol/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Transcripción Genética/genética , Activación Transcripcional , Síndrome de Williams/diagnóstico
5.
Proc Natl Acad Sci U S A ; 107(46): 19891-6, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21041627

RESUMEN

Testis-specific protein on Y chromosome (TSPY) is an ampliconic gene on the Y chromosome, and genetic interaction with gonadoblastoma has been clinically established. However, the function of the TSPY protein remains to be characterized in physiological and pathological settings. In the present study, we observed coexpression of TSPY and the androgen receptor (AR) in testicular germ-cell tumors (TGCTs) in patients as well as in model cell lines, but such coexpression was not seen in normal testis of humans or mice. TSPY was a repressor for androgen signaling because of its trapping of cytosolic AR even in the presence of androgen. Androgen treatment stimulated cell proliferation of a TGCT model cell line, and TSPY potently attenuated androgen-dependent cell growth. Together with the finding that TSPY expression is reduced in more malignant TGCTs in vivo, the present study suggests that TSPY serves as a repressor in androgen-induced tumor development in TGCTs and raises the possibility that TSPY could be used as a clinical marker to assess the malignancy of TGCTs.


Asunto(s)
Andrógenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias de Células Germinales y Embrionarias/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Testiculares/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Ciclina D2/genética , Ciclina D2/metabolismo , Citoplasma/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/patología , Unión Proteica , Transporte de Proteínas , Receptores Androgénicos/genética , Proteínas Represoras/genética , Neoplasias Testiculares/genética , Neoplasias Testiculares/patología , Transcripción Genética
6.
J Biol Chem ; 285(24): 18166-76, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20400511

RESUMEN

Estrogen-related receptor alpha (ERRalpha) is a member of the nuclear receptor superfamily and regulates many physiological functions, including mitochondrial biogenesis and lipid metabolism. ERRalpha enhances the transactivation function without endogenous ligand by associating with coactivators such as peroxisome proliferator-activated receptor gamma coactivator 1 alpha and beta (PGC-1alpha and -beta) and members of the steroid receptor coactivator family. However, the molecular mechanism by which the transactivation function of ERRalpha is converted from a repressive state to an active state is poorly understood. Here we used biochemical purification techniques to identify ERRalpha-associated proteins in HeLa cells stably expressing ERRalpha. Interestingly, we found that double PHD fingers protein DPF2/BAF45d suppressed PGC-1alpha-dependent transactivation of ERRalpha by recognizing acetylated histone H3 and associating with HDAC1. DPF2 directly bound to ERRalpha and suppressed the transactivation function of nuclear receptors such as androgen receptor. DPF2 was recruited to ERR target gene promoters in myoblast cells, and knockdown of DPF2 derepressed the level of mRNA expressed by target genes of ERRalpha. These results show that DPF2 acts as a nuclear receptor-selective co-repressor for ERRalpha by associating with both acetylated histone H3 and HDAC1.


Asunto(s)
Proteínas de Unión al ADN/química , Histona Desacetilasa 1/química , Receptores de Estrógenos/química , Proteínas Represoras/química , Acetilación , Animales , Diferenciación Celular , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Histona Desacetilasa 1/genética , Histonas/química , Humanos , Ratones , Mutación , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Factores de Transcripción , Transcripción Genética , Activación Transcripcional , Receptor Relacionado con Estrógeno ERRalfa
7.
J Bone Miner Res ; 25(5): 1157-66, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19888899

RESUMEN

Vitamin D plays an important role in regulating bone and calcium metabolism. The actions of vitamin D are mediated through the nuclear vitamin D receptor (VDR), and gene disruption of the VDR in mice causes skeletal disorders. However, the precise role of the VDR in each stage of osteoblastogenesis is not well understood. To address this issue, we used a biochemical approach to identify an osteoblast-specific coregulator of the VDR. Using a GST-fused VDR ligand-binding domain as bait, proteins associated with liganded VDR were purified from nuclear extracts of HOS osteoblastic cells and compared with those of HeLa cells. Among the interactants identified by mass fingerprinting, CCAAT displacement protein (CDP) was found as a novel ligand-dependent VDR interactant in HOS cells, together with other previously reported DRIP/TRAP complex components. Further biochemical analysis showed that complex formation between the VDR and CDP was distinct from the previously known DRIP/TRAP complex and the p160 family coactivator complexes. Transient expression of CDP potentiated VDR-mediated transcriptional activation in HOS cells. Furthermore, modulation of CDP expression levels in osteoblastic SaM-1 cells affected vitamin D-dependent osteoblast differentiation before the maturation (mineralization) stage. These findings suggest that CDP is a novel differentiation stage-specific coactivator of the VDR in osteoblasts.


Asunto(s)
Proteínas de Homeodominio/fisiología , Proteínas Nucleares/fisiología , Receptores de Calcitriol/fisiología , Proteínas Represoras/fisiología , Diferenciación Celular/efectos de los fármacos , Humanos , Osteoblastos/metabolismo , Osteosarcoma/metabolismo , Receptores de Calcitriol/genética , Factores de Transcripción , Células Tumorales Cultivadas
8.
J Biol Chem ; 284(47): 32472-82, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19776015

RESUMEN

Changes in the environment of a cell precipitate extracellular signals and sequential cascades of protein modification and elicit nuclear transcriptional responses. However, the functional links between intracellular signaling-dependent gene regulation and epigenetic regulation by chromatin-modifying proteins within the nucleus are largely unknown. Here, we describe novel epigenetic regulation by MAPK cascades that modulate formation of an ATP-dependent chromatin remodeling complex, WINAC (WSTF Including Nucleosome Assembly Complex), an SWI/SNF-type complex containing Williams syndrome transcription factor (WSTF). WSTF, a specific component of two chromatin remodeling complexes (SWI/SNF-type WINAC and ISWI-type WICH), was phosphorylated by the stimulation of MAPK cascades in vitro and in vivo. Ser-158 residue in the WAC (WSTF/Acf1/cbpq46) domain, located close to the N terminus of WSTF, was identified as a major phosphorylation target. Using biochemical analysis of a WSTF mutant (WSTF-S158A) stably expressing cell line, the phosphorylation of this residue (Ser-158) was found to be essential for maintaining the association between WSTF and core BAF complex components, thereby maintaining the ATPase activity of WINAC. WINAC-dependent transcriptional regulation of vitamin D receptor was consequently impaired by this WSTF mutation, but the recovery from DNA damage mediated by WICH was not impaired. Our results suggest that WSTF serves as a nuclear sensor of the extracellular signals to fine-tune the chromatin remodeling activity of WINAC. WINAC mediates a previously unknown MAPK-dependent step in epigenetic regulation, and this MAPK-dependent switching mechanism between the two functionally distinct WSTF-containing complexes might underlie the diverse functions of WSTF in various nuclear events.


Asunto(s)
Cromatina/química , Sistema de Señalización de MAP Quinasas , Factores de Transcripción/química , Animales , Línea Celular , Línea Celular Tumoral , Daño del ADN , Epigénesis Genética , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Mutación , Fosforilación , Estructura Terciaria de Proteína , Factores de Transcripción/metabolismo
9.
Mol Cell Biol ; 29(1): 83-92, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18981223

RESUMEN

Estrogen exerts its diverse effects through two subtypes of estrogen receptors (ER), ERalpha and ERbeta. Each subtype has its own distinct function and expression pattern in its target tissues. Little, however, is known about the transcriptional regulatory mechanism of ERbeta in the major ERbeta-expressing tissues. Using biochemical methods, we identified and described a novel ERbeta coactivator. This protein, designated GIOT-4, was biochemically purified from 293F cells. It coactivated ERbeta in ovarian granulosa cells. GIOT-4 expression was induced by stimulation with follicle-stimulating hormone (FSH). GIOT-4 recruited an SWI/SNF-type complex in a ligand-independent manner to ERbeta as an ER subtype-specific physical bridging factor and induced subsequent histone modifications in the ERbeta target gene promoters in a human ovarian granulosa cell line (KGN). Indeed, two ERbeta-specific target genes were upregulated by FSH at a specific stage of a normal ovulatory cycle in intact mice. These findings imply the presence of a novel regulatory convergence between the gonadotropin signaling cascade and ERbeta-mediated transcription in the ovary.


Asunto(s)
Receptor beta de Estrógeno/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Hormona Folículo Estimulante/análogos & derivados , Hormona Folículo Estimulante/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Gonadotropinas Equinas/farmacología , Histonas/metabolismo , Humanos , Ratones , Modelos Biológicos , Organogénesis/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/embriología , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Elementos de Respuesta , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos
10.
EMBO Rep ; 9(6): 563-8, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18451880

RESUMEN

The female sex steroid hormone oestrogen stimulates both cell proliferation and cell differentiation in target tissues. These biological actions are mediated primarily through nuclear oestrogen receptors (ERs). The ligand-dependent transactivation of ERs requires several nuclear co-regulator complexes; however, the cell-cycle-dependent associations of these complexes are poorly understood. By using a synchronization system, we found that the transactivation function of ERalpha at G2/M was lowered. Biochemical approaches showed that ERalpha associated with two discrete classes of ATP-dependent chromatin-remodelling complex in a cell-cycle-dependent manner. The components of the NuRD-type complex were identified as G2/M-phase-specific ERalpha co-repressors. Thus, our results indicate that the transactivation function of ERalpha is cell-cycle dependent and is coupled with a cell-cycle-dependent association of chromatin-remodelling complexes.


Asunto(s)
Ciclo Celular/fisiología , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Receptor alfa de Estrógeno/metabolismo , Sustancias Macromoleculares/metabolismo , Línea Celular , Receptor alfa de Estrógeno/genética , Estrógenos/metabolismo , Femenino , Humanos , Activación Transcripcional
11.
Mol Cell Biol ; 28(12): 3995-4003, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18391013

RESUMEN

TLX is an orphan nuclear receptor (also called NR2E1) that regulates the expression of target genes by functioning as a constitutive transrepressor. The physiological significance of TLX in the cytodifferentiation of neural cells in the brain is known. However, the corepressors supporting the transrepressive function of TLX have yet to be identified. In this report, Y79 retinoblastoma cells were subjected to biochemical techniques to purify proteins that interact with TLX, and we identified LSD1 (also called KDM1), which appears to form a complex with CoREST and histone deacetylase 1. LSD1 interacted with TLX directly through its SWIRM and amine oxidase domains. LSD1 potentiated the transrepressive function of TLX through its histone demethylase activity as determined by a luciferase assay using a genomically integrated reporter gene. LSD1 and TLX were recruited to a TLX-binding site in the PTEN gene promoter, accompanied by the demethylation of H3K4me2 and deacetylation of H3. Knockdown of either TLX or LSD1 derepressed expression of the endogenous PTEN gene and inhibited cell proliferation of Y79 cells. Thus, the present study suggests that LSD1 is a prime corepressor for TLX.


Asunto(s)
Regulación de la Expresión Génica , Oxidorreductasas N-Desmetilantes/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Genes Reporteros , Histona Desacetilasas/metabolismo , Histona Demetilasas , Histonas/metabolismo , Humanos , Modelos Biológicos , Receptores Nucleares Huérfanos , Oxidorreductasas N-Desmetilantes/metabolismo , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína
12.
Nat Cell Biol ; 9(11): 1273-85, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17952062

RESUMEN

Histone modifications induced by activated signalling cascades are crucial to cell-lineage decisions. Osteoblast and adipocyte differentiation from common mesenchymal stem cells is under transcriptional control by numerous factors. Although PPAR-gamma (peroxisome proliferator activated receptor-gamma) has been established as a prime inducer of adipogenesis, cellular signalling factors that determine cell lineage in bone marrow remain generally unknown. Here, we show that the non-canonical Wnt pathway through CaMKII-TAK1-TAB2-NLK transcriptionally represses PPAR-gamma transactivation and induces Runx2 expression, promoting osteoblastogenesis in preference to adipogenesis in bone marrow mesenchymal progenitors. Wnt-5a activates NLK (Nemo-like kinase), which in turn phosphorylates a histone methyltransferase, SETDB1 (SET domain bifurcated 1), leading to the formation of a co-repressor complex that inactivates PPAR-gamma function through histone H3-K9 methylation. These findings suggest that the non-canonical Wnt signalling pathway suppresses PPAR-gamma function through chromatin inactivation triggered by recruitment of a repressing histone methyltransferase, thus leading to an osteoblastic cell lineage from mesenchymal stem cells.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , PPAR gamma/metabolismo , Transducción de Señal/fisiología , Activación Transcripcional/fisiología , Proteínas Wnt/fisiología , Adipogénesis , Animales , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación hacia Abajo , Vectores Genéticos , N-Metiltransferasa de Histona-Lisina/efectos de los fármacos , Ratones , Ratones Transgénicos , Mutación , Osteogénesis , PPAR gamma/efectos de los fármacos , PPAR gamma/genética , Fosforilación , Plásmidos , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/genética , Proteínas Wnt/farmacología , Proteína Wnt-5a
13.
Mol Cell Biol ; 27(22): 7947-54, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17875939

RESUMEN

Vitamin K is a fat-soluble vitamin that serves as a coenzyme for vitamin K-dependent carboxylase. Besides its canonical action, vitamin K binds to the steroid and xenobiotic receptor (SXR)/pregnane X receptor (PXR) and modulates gene transcription. To determine if the osteoprotective action of vitamin K is the result of the PXR/SXR pathway, we screened by two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis the PXR/SXR target genes in an osteoblastic cell line (MC3T3-E1) treated with a vitamin K2 (menaquinone 4 [MK4]). Osteoblastic differentiation of MC3T3-E1 cells was induced by MK4. Msx2, an osteoblastogenic transcription factor, was identified as an MK4-induced gene. Functional analysis of the Msx2 gene promoter mapped a vitamin K-responsive element (PXR-responsive element [PXRE]) that was directly bound by a PXR/retinoid X receptor alpha heterodimer. In a chromatin immunoprecipitation analysis, PXR was recruited together with a coactivator, p300, to the PXRE in the Msx2 promoter. MK4-bound PXR cooperated with estrogen-bound estrogen receptor alpha to control transcription at the Msx2 promoter. Knockdown of either PXR or Msx2 attenuated the effect of MK4 on osteoblastic differentiation. Thus, the present study suggests that Msx2 is a target gene for PXR activated by vitamin K and suggests that the osteoprotective action of MK4 in the human mediates, at least in part, a genomic pathway of vitamin K signaling.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Proteínas de Homeodominio , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Receptores de Esteroides/metabolismo , Vitamina K/farmacología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Hemostáticos/metabolismo , Hemostáticos/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Noqueados , Osteoblastos/citología , Receptor X de Pregnano , Regiones Promotoras Genéticas , Receptores de Esteroides/genética , Elementos de Respuesta , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Transducción de Señal/fisiología , Transcripción Genética , Vitamina K/metabolismo , Vitamina K 2/análogos & derivados , Vitamina K 2/metabolismo , Vitamina K 2/farmacología
14.
Mol Endocrinol ; 21(2): 334-42, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17095575

RESUMEN

Although transactivation by the liganded vitamin D receptor (VDR) is well described at the molecular level, the precise molecular mechanism of negative regulation by the liganded VDR remains to be elucidated. We have previously reported a novel class of negative vitamin D response element (nVDRE) called 1alphanVDRE in the human 25(OH)D31alpha-hydroxylase [1alpha(OH)ase] gene by 1alpha,25(OH)2D3-bound VDR. This element was composed of two E-box-type motifs that bound to VDIR for transactivation, which was attenuated by liganded VDR. Here, we explore the possible functions of VDIR and E-box motifs in the human (h) PTH and hPTHrP gene promoters. Functional mapping of the hPTH and hPTHrP promoters identified E-box-type elements acting as nVDREs in both the hPTH promoter (hPTHnVDRE; -87 to -60 bp) and in the hPTHrP promoter (hPTHrPnVDRE; -850 to -600 bp; -463 to -104 bp) in a mouse renal tubule cell line. The hPTHnVDRE alone was enough to direct ligand-induced transrepression mediated through VDR/retinoid X receptor and VDIR. Direct DNA binding of hPTHnVDRE to VDIR, but not VDR/retinoid X receptor, was observed and ligand-induced transrepression was coupled with recruitment of VDR and histone deacetylase 2 (HDAC2) to the hPTH promoter. These results suggest that negative regulation of the hPTH gene by liganded VDR is mediated by VDIR directly binding to the E-box-type nVDRE at the promoter, together with recruitment of an HDAC corepressor for ligand-induced transrepression.


Asunto(s)
Calcitriol/farmacología , Elementos E-Box , Hormona Paratiroidea/fisiología , Regiones Promotoras Genéticas , Receptores de Calcitriol/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Humanos , Ratones , Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Receptores de Calcitriol/genética , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Elemento de Respuesta a la Vitamina D
15.
J Biol Chem ; 281(1): 20-6, 2006 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-16260778

RESUMEN

It is well established that genetic mutations that impair BRCA1 function predispose women to early onset of breast and ovarian cancer. However, the co-regulatory factors that support normal BRCA1 functions remain to be identified. Using a biochemical approach to search for such co-regulatory factors, we identified hGCN5, TRRAP, and hMSH2/6 as BRCA1-interacting proteins. Genetic mutations in the C-terminal transactivation domain of BRCA1, as found in breast cancer patients (Chapman, M. S., and Verma, I. M. (1996) Nature 382, 678-679), caused the loss of physical interaction between BRCA1 and TRRAP and significantly reduced the co-activation of BRCA1 transactivation function by hGCN5/TRRAP. The reported transcriptional squelching between BRCA1 and estrogen receptor alpha (Fan, S., Wang, J., Yuan, R., Ma, Y., Meng, Q., Erdos, M. R., Pestell, R. G., Yuan, F., Auborn, K. J., Goldberg, I. D., and Rosen, E. M. (1999) Science 284, 1354-1356) was rescued by the overexpression of TRRAP or hGCN5. Histone acetyltransferase hGCN5 activity appeared to be indispensable for coregulator complex function in both BRCA1-mediated gene regulation and DNA repair. Biochemical purification of the hGCN5/TRRAP-containing complex suggested that hGCN5/TRRAP formed a complex with hMSH2/hMSH6, presumably as a novel subclass of hGCN5/TRRAP-containing known TFTC (TBP-free TAF-containing)-type histone acetyltransferase complex (hTFTC, hPCAF, and hSTAGA) (Yanagisawa, J., Kitagawa, H., Yanagida, M., Wada, O., Ogawa, S., Nakagomi, M., Oishi, H., Yamamoto, Y., Nagasawa, H., McMahon, S. B., Cole, M. D., Tora, L., Takahashi, N., and Kato, S. (2002) Mol. Cell 9, 553-562). Unlike other subclasses, the isolated complex harbored a previously unknown combination of components including hMSH2 and hMSH6, major components of the BRCA1 genome surveillance repair complex (BASC). Thus, our results suggested that the multiple BRCA1 functions require a novel hGCN5/TRRAP histone acetyltransferase complex subclass.


Asunto(s)
Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteína BRCA1/genética , Neoplasias de la Mama/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Histona Acetiltransferasas/genética , Humanos , Complejos Multienzimáticos/metabolismo , Proteínas Nucleares/genética , Factores de Transcripción/genética , Transcripción Genética/fisiología , Factores de Transcripción p300-CBP
16.
Genes Cells ; 10(12): 1095-102, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16324147

RESUMEN

Estrogen receptor (ER) is a hormone-inducible transcription factor as a member of the nuclear receptor gene superfamily. Unliganded ER is transcriptionally silent and capable of DNA binding; however, it is unable to suppress the basal activity of the target gene promoters, unlike non-steroid hormone receptors that associate with corepressors in the absence of their cognate ligands. To study the molecular basis of how unliganded human ERalpha is maintained silent in gene regulation upon the target gene promoters, we biochemically searched interactants for hERalpha, and identified heat shock protein 70 (Hsc70). Hsc70 appeared to associate with the N-terminal hormone binding E domain, that also turned out a transcriptionally repressive domain. Competitive association of Hsc70 with a best known coactivator p300 was observed. Thus, these findings suggest that Hsc70 associates with unliganded hERalpha, and thereby deters hERalpha from recruiting transcriptional coregulators, presumably as a component of chaperone complexes.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Ligandos , Proteínas Represoras/metabolismo , Células Cultivadas , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/genética , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Células HeLa , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica/genética , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Transcripción Genética , Activación Transcripcional/genética , Transfección
17.
Genes Cells ; 9(2): 143-51, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15009097

RESUMEN

Smad proteins are intracellular signalling mediators of transforming growth factor-beta (TGF-beta) superfamily. In the nucleus, activated Smad complexes regulate transcriptional responses of the target genes in cooperation with transcriptional coactivators and corepressors. To identify new components of transcriptional complexes containing Smad proteins, we purified DNA-binding proteins from human breast cancer MCF-7 cell nuclear extract using a Smad-binding DNA element as bait, and identified a coactivator GCN5 as a direct partner of activated Smad complexes. GCN5 is structurally similar to PCAF, which was previously identified as a coactivator for receptor-regulated Smads (R-Smads) for TGF-beta signalling pathways. GCN5 functions like PCAF, in that it binds to TGF-beta-specific R-Smads, and enhances transcriptional activity induced by TGF-beta. In addition, GCN5, but not PCAF, interacts with R-Smads for bone morphogenetic protein (BMP) signalling pathways, and enhances BMP-induced transcriptional activity, suggesting that GCN5 and PCAF have distinct physiological functions in vivo. Moreover, silencing of the GCN5 gene by RNA interference results in repression of transcriptional activities induced by TGF-beta. In conclusion we identified GCN5 as a Smad-binding transcriptional coactivator which positively regulates both TGF-beta and BMP signalling pathways.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Acetiltransferasas/metabolismo , Animales , Sitios de Unión , Receptores de Proteínas Morfogenéticas Óseas , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular , Extractos Celulares , Línea Celular , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Histona Acetiltransferasas , Humanos , Inhibidor 1 de Activador Plasminogénico/genética , Pruebas de Precipitina , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Secuencias Reguladoras de Ácidos Nucleicos/fisiología , Transducción de Señal , Proteínas Smad , Proteína smad3 , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología , Factores de Transcripción p300-CBP
19.
Biochem Biophys Res Commun ; 306(3): 660-5, 2003 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-12810069

RESUMEN

The androgen receptor (AR) has two transactivation functions that have been mapped to the N- and C-terminal domains and designated as activation function-1 (AF-1) and AF-2, respectively. While the molecular basis for AF-2 function has been well studied, little is known about AF-1 coregulators. Therefore, we attempted to identify AF-1-interacting proteins from HEK293 cells by biochemical purification followed by mass fingerprinting by matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (MALDI-TOF MS). Purified AF-1 region-interacting proteins were found to contain nuclear RNA-binding protein p54(nrb), polypyrimidine tract-binding protein-associated splicing factor (PSF), paraspeckle protein 1 (PSP1), and PSP2, which are assumed to be involved in pre-mRNA processing. p54(nrb) interacted with AR via the A/B domain in a ligand-dependent manner. Reflecting the physical interaction between p54(nrb) and the AR A/B domain, AR AF-1 function was potentiated by p54(nrb). Our results suggest that p54(nrb) functions as a coactivator of AR that potentiates transcription, and presumably splicing as well.


Asunto(s)
Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores Androgénicos/metabolismo , Activación Transcripcional , Línea Celular , Proteínas de Unión al ADN , Genes Reporteros , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción de Octámeros , Mapeo Peptídico , Unión Proteica , Estructura Terciaria de Proteína , Receptores Androgénicos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Nature ; 423(6939): 545-50, 2003 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-12774124

RESUMEN

Environmental contaminants affect a wide variety of biological events in many species. Dioxins are typical environmental contaminants that exert adverse oestrogen-related effects. Although their anti-oestrogenic actions are well described, dioxins can also induce endometriosis and oestrogen-dependent tumours, implying possible oestrogenic effects. However, the molecular mechanism underlying oestrogen-related actions of dioxins remains largely unknown. A heterodimer of the dioxin receptor (AhR) and Arnt, which are basic helix-loop-helix/PAS-family transcription factors, mediates most of the toxic effects of dioxins. Here we show that the agonist-activated AhR/Arnt heterodimer directly associates with oestrogen receptors ER-alpha and ER-beta. This association results in the recruitment of unliganded ER and the co-activator p300 to oestrogen-responsive gene promoters, leading to activation of transcription and oestrogenic effects. The function of liganded ER is attenuated. Oestrogenic actions of AhR agonists were detected in wild-type ovariectomized mouse uteri, but were absent in AhR-/- or ER-alpha-/- ovariectomized mice. Our findings suggest a novel mechanism by which ER-mediated oestrogen signalling is modulated by a co-regulatory-like function of activated AhR/Arnt, giving rise to adverse oestrogen-related actions of dioxin-type environmental contaminants.


Asunto(s)
Proteínas de Unión al ADN , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo , Sitios de Unión , Células COS , Dioxinas/farmacología , Contaminantes Ambientales/farmacología , Estrógenos/farmacología , Femenino , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Metilcolantreno/farmacología , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/genética , Receptores de Estrógenos/química , Receptores de Estrógenos/genética , Elementos de Respuesta/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/química , Factores de Transcripción/genética , Células Tumorales Cultivadas , Útero/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA