Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Physiol Rev ; 93(2): 481-523, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23589826

RESUMEN

During the last decade, our view on the skeleton as a mere solid physical support structure has been transformed, as bone emerged as a dynamic, constantly remodeling tissue with systemic regulatory functions including those of an endocrine organ. Reflecting this remarkable functional complexity, distinct classes of humoral and intracellular regulatory factors have been shown to control vital processes in the bone. Among these regulators, nuclear receptors (NRs) play fundamental roles in bone development, growth, and maintenance. NRs are DNA-binding transcription factors that act as intracellular transducers of the respective ligand signaling pathways through modulation of expression of specific sets of cognate target genes. Aberrant NR signaling caused by receptor or ligand deficiency may profoundly affect bone health and compromise skeletal functions. Ligand dependency of NR action underlies a major strategy of therapeutic intervention to correct aberrant NR signaling, and significant efforts have been made to design novel synthetic NR ligands with enhanced beneficial properties and reduced potential negative side effects. As an example, estrogen deficiency causes bone loss and leads to development of osteoporosis, the most prevalent skeletal disorder in postmenopausal women. Since administration of natural estrogens for the treatment of osteoporosis often associates with undesirable side effects, several synthetic estrogen receptor ligands have been developed with higher therapeutic efficacy and specificity. This review presents current progress in our understanding of the roles of various nuclear receptor-mediated signaling pathways in bone physiology and disease, and in development of advanced NR ligands for treatment of common skeletal disorders.


Asunto(s)
Desarrollo Óseo/genética , Huesos/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología , Animales , Desarrollo Óseo/fisiología , Huesos/citología , Huesos/fisiopatología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Humanos , Ligandos
2.
Blood ; 124(25): 3791-8, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25270909

RESUMEN

Diamond-Blackfan anemia (DBA) is an inherited disorder characterized by defects in erythropoiesis, congenital abnormalities, and predisposition to cancer. Approximately 25% of DBA patients have a mutation in RPS19, which encodes a component of the 40S ribosomal subunit. Upregulation of p53 contributes to the pathogenesis of DBA, but the link between ribosomal protein mutations and erythropoietic defects is not well understood. We found that RPS19 deficiency in hematopoietic progenitor cells leads to decreased GATA1 expression in the erythroid progenitor population and p53-dependent upregulation of tumor necrosis factor-α (TNF-α) in nonerythroid cells. The decrease in GATA1 expression was mediated, at least in part, by activation of p38 MAPK in erythroid cells and rescued by inhibition of TNF-α or p53. The anemia phenotype in rps19-deficient zebrafish was reversed by treatment with the TNF-α inhibitor etanercept. Our data reveal that RPS19 deficiency leads to inflammation, p53-dependent increase in TNF-α, activation of p38 MAPK, and decreased GATA1 expression, suggesting a novel mechanism for the erythroid defects observed in DBA.


Asunto(s)
Factor de Transcripción GATA1/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inflamación/metabolismo , Proteínas Ribosómicas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Western Blotting , Células Cultivadas , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Activación Enzimática , Células Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Etanercept , Factor de Transcripción GATA1/genética , Expresión Génica , Humanos , Inmunoglobulina G/farmacología , Inflamación/genética , Interferencia de ARN , Receptores del Factor de Necrosis Tumoral , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ribosómicas/genética , Factor de Necrosis Tumoral alfa/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
3.
Nature ; 461(7266): 1007-12, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19829383

RESUMEN

Epigenetic modifications at the histone level affect gene regulation in response to extracellular signals. However, regulated epigenetic modifications at the DNA level, especially active DNA demethylation, in gene activation are not well understood. Here we report that DNA methylation/demethylation is hormonally switched to control transcription of the cytochrome p450 27B1 (CYP27B1) gene. Reflecting vitamin-D-mediated transrepression of the CYP27B1 gene by the negative vitamin D response element (nVDRE), methylation of CpG sites ((5m)CpG) is induced by vitamin D in this gene promoter. Conversely, treatment with parathyroid hormone, a hormone known to activate the CYP27B1 gene, induces active demethylation of the (5m)CpG sites in this promoter. Biochemical purification of a complex associated with the nVDRE-binding protein (VDIR, also known as TCF3) identified two DNA methyltransferases, DNMT1 and DNMT3B, for methylation of CpG sites, as well as a DNA glycosylase, MBD4 (ref. 10). Protein-kinase-C-phosphorylated MBD4 by parathyroid hormone stimulation promotes incision of methylated DNA through glycosylase activity, and a base-excision repair process seems to complete DNA demethylation in the MBD4-bound promoter. Such parathyroid-hormone-induced DNA demethylation and subsequent transcriptional derepression are impaired in Mbd4(-/-) mice. Thus, the present findings suggest that methylation switching at the DNA level contributes to the hormonal control of transcription.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Hormona Paratiroidea/farmacología , Transcripción Genética/efectos de los fármacos , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Animales , Línea Celular , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Glicosilasas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Endodesoxirribonucleasas/deficiencia , Endodesoxirribonucleasas/genética , Ratones , Fosforilación , Proteína Quinasa C/metabolismo , Elementos de Respuesta/genética , Vitamina D/farmacología , ADN Metiltransferasa 3B
4.
Nucleic Acids Res ; 41(8): 4495-506, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23502002

RESUMEN

To determine the functional specificity of inflammation, it is critical to orchestrate the timely activation and repression of inflammatory responses. Here, we explored the PAF1 (RNA polymerase II associated factor)-mediated signal- and locus-specific repression of genes induced through the pro-inflammatory cytokine interleukin (IL)-1ß. Using microarray analysis, we identified the PAF1 target genes whose expression was further enhanced by PAF1 knockdown in IL-1ß-stimulated HepG2 hepatocarcinomas. PAF1 bound near the transcription start sites of target genes and dissociated on stimulation. In PAF1-deficient cells, more elongating RNA polymerase II and acetylated histones were observed, although IL-1ß-mediated activation and recruitment of nuclear factor κB (NF-κB) were not altered. Under basal conditions, PAF1 blocked histone acetyltransferase general control non-depressible 5 (GCN5)-mediated acetylation on H3K9 and H4K5 residues. On IL-1ß stimulation, activated GCN5 discharged PAF1 from chromatin, allowing productive transcription to occur. PAF1 bound to histones but not to acetylated histones, and the chromatin-binding domain of PAF1 was essential for target gene repression. Moreover, IL-1ß-induced cell migration was similarly controlled through counteraction between PAF1 and GCN5. These results suggest that the IL-1ß signal-specific exchange of PAF1 and GCN5 on the target locus limits inappropriate gene induction and facilitates the timely activation of inflammatory responses.


Asunto(s)
Proteínas Portadoras/metabolismo , Cromatina/metabolismo , Regulación de la Expresión Génica , Histona Acetiltransferasas/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1beta/farmacología , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Proteínas Portadoras/química , Línea Celular Tumoral , Movimiento Celular , Histonas/metabolismo , Humanos , Ratones , FN-kappa B/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , Elongación de la Transcripción Genética , Factores de Transcripción , Sitio de Iniciación de la Transcripción , Transcripción Genética , Activador de Plasminógeno de Tipo Uroquinasa/genética
5.
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
6.
J Biol Chem ; 287(16): 12994-3004, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22375008

RESUMEN

Osteoclastogenesis is a highly regulated process governed by diverse classes of regulators. Among them, nuclear factor of activated T-cells calcineurin-dependent 1 (NFATc1) is the primary osteoclastogenic transcription factor, and its expression is transcriptionally induced during early osteoclastogenesis by receptor activation of nuclear factor κB ligand (RANKL), an osteoclastogenic cytokine. Here, we report the novel enzymatic function of JMJD5, which regulates NFATc1 protein stability. Among the tested Jumonji C (JmjC) domain-containing proteins, decreased mRNA expression levels during osteoclastogenesis were found for JMJD5 in RAW264 cells stimulated by RANKL. To examine the functional role of JMJD5 in osteoclast differentiation, we established stable JMJD5 knockdown cells, and osteoclast formation was assessed. Down-regulated expression of JMJD5 led to accelerated osteoclast formation together with induction of several osteoclast-specific genes such as Ctsk and DC-STAMP, suggesting that JMJD5 is a negative regulator in osteoclast differentiation. Although JMJD5 was recently reported as a histone demethylase for histone H3K36me2, no histone demethylase activity was detected in JMJD5 in vitro or in living cells, even for other methylated histone residues. Instead, JMJD5 co-repressed transcriptional activity by destabilizing NFATc1 protein. Protein hydroxylase activity mediated by the JmjC domain in JMJD5 was required for the observed functions of JMJD5. JMJD5 induced the association of hydroxylated NFATc1 with the E3 ubiquitin ligase Von Hippel-Lindau tumor suppressor (VHL), thereby presumably facilitating proteasomal degradation of NFATc1 via ubiquitination. Taken together, the present study demonstrated that JMJD5 is a post-translational co-repressor for NFATc1 that attenuates osteoclastogenesis.


Asunto(s)
Histona Demetilasas/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Osteoclastos/enzimología , Diferenciación Celular/fisiología , Activación Enzimática/fisiología , Epigénesis Genética/fisiología , Células HEK293 , Histona Demetilasas/genética , Humanos , Hidroxilación/fisiología , Factores de Transcripción NFATC/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Activación Transcripcional/fisiología , Ubiquitina/metabolismo
8.
Clin Calcium ; 22(5): 611-7, 2012 May.
Artículo en Japonés | MEDLINE | ID: mdl-22549184

RESUMEN

Osteoclasts are differentiated from hematopoietic stem cells and become multinucleated giant cells through cell-fusion by a number of regulators. Among such regulators, transcription factors play pivotal roles by reorganizing gene networks. Recently, epigenetic regulators like histone modifiers and chromatin remodelers have emerged to be prerequisite for gene regulations by transcriptional factors. However, little is known about epigenetic controls during osteoclastogenesis and osteoclastic maturation. To address this issue, we tried to identify novel epigenetic regulators for fine control of NFATc1 function through biochemical approaches. Here, we summarize the new epigenetic regulation mechanism and epigenetic regulator which are required for normal osteoclastogenesis.


Asunto(s)
Diferenciación Celular/genética , Epigénesis Genética , Factores de Transcripción NFATC/genética , Osteoclastos/citología , Animales , Diferenciación Celular/fisiología , Ratones , Osteoclastos/fisiología
9.
Genes Cells ; 15(10): 1025-35, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20831632

RESUMEN

Multinucleation is indispensable for the bone-resorbing activity of mature osteoclasts. Although multinucleation is evident in mature osteoclasts and certain other cell types, putative regulatory networks among nuclei remain poorly characterized. To address this issue, transcriptional activity of each nucleus in a multinucleated osteoclast was assessed by detecting the distributions of nuclear proteins by immunocytochemistry and primary transcripts by RNA FISH. Patterns of epigenetic histone markers governing transcription as well as localization of tested nuclear receptor proteins appeared indistinguishable among nuclei in differentiated Raw264 cells and mouse mature osteoclasts. However, RNAPII-Ser5P/2P and NFATc1 proteins were selectively distributed in certain nuclei in the same cell. Similarly, the distributions of primary transcripts for osteoclast-specific genes (Nfatc1, Ctsk and Acp5) as well as a housekeeping gene (beta-tubulin) were limited in certain nuclei within individual cells. By fusing two Raw264 cell lines that stably expressed ZsGreen-NLS and DsRed-NLS proteins, transmission of nuclear proteins across all of the nuclei in a cell could be observed, presumably through the shared cytoplasm. Taken together, we conclude that although nuclear proteins are diffusible among nuclei, only certain nuclei within a multinucleated osteoclast are transcriptionally active.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Nucleares/metabolismo , Osteoclastos/metabolismo , Osteoclastos/ultraestructura , Transcripción Genética , Animales , Diferenciación Celular , Línea Celular , Epigenómica , Células Gigantes/citología , Células Gigantes/metabolismo , Células Gigantes/ultraestructura , Hibridación Fluorescente in Situ , Ratones , Proteínas Nucleares/genética , Osteoclastos/citología , ARN Mensajero/biosíntesis , ARN Mensajero/genética
10.
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
11.
Biosci Biotechnol Biochem ; 74(9): 1956-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20834143

RESUMEN

Multinucleation is indispensable to the bone-resorbing activity of mature osteoclasts. Nevertheless, little is known about the regulatory networks among multi-nuclei in a single mature osteoclast. For this reason, we purified osteoclastic factors from the nuclear envelope by two-dimensional gel electrophoresis. Two annexin family proteins and ferritin light chain 1 protein were identified as osteoclastic candidates.


Asunto(s)
Anexinas/análisis , Apoferritinas/análisis , Resorción Ósea/etiología , Proteínas Portadoras/análisis , Membrana Nuclear/química , Osteoclastos/química , Animales , Línea Celular , Células Cultivadas , Electroforesis en Gel Bidimensional , Redes Reguladoras de Genes , Macrófagos , Ratones
13.
Sci Rep ; 8(1): 5165, 2018 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-29581525

RESUMEN

Deficiency of ribosomal proteins (RPs) leads to Diamond Blackfan Anemia (DBA) associated with anemia, congenital defects, and cancer. While p53 activation is responsible for many features of DBA, the role of immune system is less defined. The Innate immune system can be activated by endogenous nucleic acids from non-processed pre-rRNAs, DNA damage, and apoptosis that occurs in DBA. Recognition by toll like receptors (TLRs) and Mda5-like sensors induces interferons (IFNs) and inflammation. Dying cells can also activate complement system. Therefore we analyzed the status of these pathways in RP-deficient zebrafish and found upregulation of interferon, inflammatory cytokines and mediators, and complement. We also found upregulation of receptors signaling to IFNs including Mda5, Tlr3, and Tlr9. TGFb family member activin was also upregulated in RP-deficient zebrafish and in RPS19-deficient human cells, which include a lymphoid cell line from a DBA patient, and fetal liver cells and K562 cells transduced with RPS19 shRNA. Treatment of RP-deficient zebrafish with a TLR3 inhibitor decreased IFNs activation, acute phase response, and apoptosis and improved their hematopoiesis and morphology. Inhibitors of complement and activin also had beneficial effects. Our studies suggest that innate immune system contributes to the phenotype of RPS19-deficient zebrafish and human cells.


Asunto(s)
Anemia de Diamond-Blackfan/inmunología , Anemia de Diamond-Blackfan/metabolismo , Inmunidad Innata/fisiología , Pez Cebra/inmunología , Pez Cebra/metabolismo , Receptores de Activinas/antagonistas & inhibidores , Activinas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Benzamidas/farmacología , Compuestos de Bencidrilo/farmacología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/metabolismo , Dioxoles/farmacología , Modelos Animales de Enfermedad , Humanos , Interferones/metabolismo , Células K562 , ARN Interferente Pequeño/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Proteínas Ribosómicas/metabolismo , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo
14.
Ann N Y Acad Sci ; 1192: 201-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20392237

RESUMEN

Mature osteoclasts are multinuclear, macrophage-like cells derived from hematopoietic stem cells in the bone marrow. Several transcription factors regulating osteoclast differentiation have been identified. However, the molecular basis of transcriptional regulation in osteoclasts at epigenetic levels is largely unknown. In fact, no osteoclast-specific transcriptional co-regulators have been characterized. Recently, selective ablation of estrogen receptor alpha (ERalpha) in mature osteoclasts derived from female mice (ERalpha(Deltaoc/Deltaoc)) exhibited trabecular bone loss due to induced apoptosis via upregulated expression of Fas ligand mRNA. In general, the component composition of the ERalpha-associated co-activator complex and its expression levels are distinct among tissues. However, ERalpha transcriptional co-regulators in mature osteoclasts remain unclear. In the present study, we achieved large-scale cultivation of mature, multinucleated osteoclasts and established a purification system for ERalpha-associated proteins. In addition to co-regulators previously found in other ERalpha target cells, several unexpected factors were found such as CAP-H. The mRNA expression level of CAP-H was high during osteoclast differentiation. These results demonstrate the existence of osteoclast-specific transcriptional co-regulators supporting ERalpha function.


Asunto(s)
Proteínas Co-Represoras/aislamiento & purificación , Receptor alfa de Estrógeno/metabolismo , Osteoclastos/metabolismo , Transactivadores/aislamiento & purificación , Animales , Clonación Molecular , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Receptor alfa de Estrógeno/aislamiento & purificación , Femenino , Humanos , Ratones , Modelos Biológicos , Osteoclastos/química , Transactivadores/genética , Transactivadores/metabolismo , Activación Transcripcional/genética
15.
J Biol Chem ; 282(48): 34727-34, 2007 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-17911113

RESUMEN

PAF, which is composed of Paf1, Cdc73, Ctr9, Leo1, and Rtf1, is a novel complex with multiple functions in transcription-related activities. The PAF complex interacts with histone-modifying enzymes and RNA polymerase II to regulate transcription. With general transcription regulatory potential in yeast, Hyrax/Cdc73 has been reported to associate with beta-catenin to control Wnt/Wg signal-specific transcription in Drosophila. Here, we present the first evidence of IL-6 signal-specific transcriptional regulation by SH2BP1/CTR9 in mammals. Upon LPS injection of mice, we observed transient induction of the mammalian PAF complex in the liver. Inhibition of CTR9 specifically abrogated expression of IL-6-responsive genes, but had no effect on genes constitutively expressed or induced by interferon-beta, TNFalpha, or IL-1beta. The PAF complex was found in the promoter regions of IL-6-responsive HP and FGGgamma, but not in the promoter region of constitutively active GAPDH. Transcriptional activation by STAT3 was inhibited when CTR9 siRNA was introduced, whereas transcriptional activation was enhanced by mCtr9 overexpression. IL-6-activated Stat3 was found to co-localize and interact with CTR9. In CTR9-depleted cells, decreased STAT3 association with the promoter regions, as well as impaired K4-trimethylation of histone H3 in the coding regions, of target genes was observed. These data suggest that CTR9 participates in the transcription of IL-6-responsive genes through the regulation of DNA association of STAT3 and modification of histone methylation.


Asunto(s)
ADN/metabolismo , Regulación de la Expresión Génica , Interleucina-6/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transcripción Genética , Animales , Células COS , Chlorocebus aethiops , Metilación de ADN , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA