Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Nature ; 480(7378): 557-60, 2011 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-22121020

RESUMO

Chromatin reorganization is governed by multiple post-translational modifications of chromosomal proteins and DNA. These histone modifications are reversible, dynamic events that can regulate DNA-driven cellular processes. However, the molecular mechanisms that coordinate histone modification patterns remain largely unknown. In metazoans, reversible protein modification by O-linked N-acetylglucosamine (GlcNAc) is catalysed by two enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). However, the significance of GlcNAcylation in chromatin reorganization remains elusive. Here we report that histone H2B is GlcNAcylated at residue S112 by OGT in vitro and in living cells. Histone GlcNAcylation fluctuated in response to extracellular glucose through the hexosamine biosynthesis pathway (HBP). H2B S112 GlcNAcylation promotes K120 monoubiquitination, in which the GlcNAc moiety can serve as an anchor for a histone H2B ubiquitin ligase. H2B S112 GlcNAc was localized to euchromatic areas on fly polytene chromosomes. In a genome-wide analysis, H2B S112 GlcNAcylation sites were observed widely distributed over chromosomes including transcribed gene loci, with some sites co-localizing with H2B K120 monoubiquitination. These findings suggest that H2B S112 GlcNAcylation is a histone modification that facilitates H2BK120 monoubiquitination, presumably for transcriptional activation.


Assuntos
Acetilglucosamina/metabolismo , Histonas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Células HeLa , Histonas/química , Histonas/genética , Humanos , Modelos Moleculares , Mutação , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ubiquitinação
2.
Nature ; 459(7245): 455-9, 2009 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-19377461

RESUMO

The post-translational modifications of histone tails generate a 'histone code' that defines local and global chromatin states. The resultant regulation of gene function is thought to govern cell fate, proliferation and differentiation. Reversible histone modifications such as methylation are under mutual controls to organize chromosomal events. Among the histone modifications, methylation of specific lysine and arginine residues seems to be critical for chromatin configuration and control of gene expression. Methylation of histone H3 lysine 4 (H3K4) changes chromatin into a transcriptionally active state. Reversible modification of proteins by beta-N-acetylglucosamine (O-GlcNAc) in response to serum glucose levels regulates diverse cellular processes. However, the epigenetic impact of protein GlcNAcylation is unknown. Here we report that nuclear GlcNAcylation of a histone lysine methyltransferase (HKMT), MLL5, by O-GlcNAc transferase facilitates retinoic-acid-induced granulopoiesis in human HL60 promyelocytes through methylation of H3K4. MLL5 is biochemically identified in a GlcNAcylation-dependent multi-subunit complex associating with nuclear retinoic acid receptor RARalpha (also known as RARA), serving as a mono- and di-methyl transferase to H3K4. GlcNAcylation at Thr 440 in the MLL5 SET domain evokes its H3K4 HKMT activity and co-activates RARalpha in target gene promoters. Increased nuclear GlcNAcylation by means of O-GlcNAc transferase potentiates retinoic-acid-induced HL60 granulopoiesis and restores the retinoic acid response in the retinoic-acid-resistant HL60-R2 cell line. Thus, nuclear MLL5 GlcNAcylation triggers cell lineage determination of HL60 through activation of its HKMT activity.


Assuntos
Acetilglucosamina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Granulócitos/citologia , Granulócitos/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Leucopoese/efeitos dos fármacos , N-Acetilglucosaminiltransferases/metabolismo , Tretinoína/farmacologia , Linhagem da Célula , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Células HL-60 , Histona-Lisina N-Metiltransferase/química , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/isolamento & purificação , Complexos Multiproteicos/metabolismo , N-Acetilglucosaminiltransferases/química , Estrutura Terciária de Proteína , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Treonina/metabolismo
3.
Nature ; 461(7266): 1007-12, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19829383

RESUMO

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.


Assuntos
Metilação de DNA/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Transcrição Gênica/efeitos dos fármacos , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , Animais , Linhagem Celular , Ilhas de CpG/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Glicosilases/metabolismo , Regulação para Baixo/efeitos dos fármacos , Endodesoxirribonucleases/deficiência , Endodesoxirribonucleases/genética , Camundongos , Fosforilação , Proteína Quinase C/metabolismo , Elementos de Resposta/genética , Vitamina D/farmacologia , DNA Metiltransferase 3B
4.
Nat Cell Biol ; 9(11): 1273-85, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17952062

RESUMO

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.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , PPAR gama/metabolismo , Transdução de Sinais/fisiologia , Ativação Transcricional/fisiologia , Proteínas Wnt/fisiologia , Adipogenia , Animais , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação para Baixo , Vetores Genéticos , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Mutação , Osteogênese , PPAR gama/efeitos dos fármacos , PPAR gama/genética , Fosforilação , Plasmídeos , Transdução de Sinais/efeitos dos fármacos , Proteínas Wnt/genética , Proteínas Wnt/farmacologia , Proteína Wnt-5a
5.
Nat Cell Biol ; 9(5): 604-11, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17435748

RESUMO

MicroRNAs (miRNAs) control cell proliferation, differentiation and fate through modulation of gene expression by partially base-pairing with target mRNA sequences. Drosha is an RNase III enzyme that is the catalytic subunit of a large complex that cleaves pri-miRNAs with distinct structures into pre-miRNAs. Here, we show that both the p68 and p72 DEAD-box RNA helicase subunits in the mouse Drosha complex are indispensable for survival in mice, and both are required for primary miRNA and rRNA processing. Gene disruption of either p68 or p72 in mice resulted in early lethality, and in both p68(-/-) and p72(-/-) embryos, expression levels of a set of, but not all, miRNAs and 5.8S rRNA were significantly lowered. In p72(-/-) MEF cells, expression of p72, but not a mutant lacking ATPase activity, restored the impaired expression of miRNAs and 5.8S rRNA. Furthermore, we purified the large complex of mouse Drosha and showed it could generate pre-miRNA and 5.8S rRNA in vitro. Thus, we suggest that DEAD-box RNA helicase subunits are required for recognition of a subset of primary miRNAs in mDrosha-mediated processing.


Assuntos
RNA Helicases DEAD-box/metabolismo , Embrião de Mamíferos/metabolismo , MicroRNAs/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA , RNA Ribossômico/metabolismo , Ribonuclease III/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Imunoprecipitação da Cromatina , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , RNA Ribossômico 5,8S/metabolismo
7.
Proc Natl Acad Sci U S A ; 107(46): 19891-6, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21041627

RESUMO

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.


Assuntos
Androgênios/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias Embrionárias de Células Germinativas/metabolismo , Receptores Androgênicos/metabolismo , Proteínas Repressoras/metabolismo , Neoplasias Testiculares/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D2/genética , Ciclina D2/metabolismo , Citoplasma/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Ligação Proteica , Transporte Proteico , Receptores Androgênicos/genética , Proteínas Repressoras/genética , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia , Transcrição Gênica
10.
Proc Natl Acad Sci U S A ; 106(23): 9280-5, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19470456

RESUMO

A number of nuclear complexes modify chromatin structure and operate as functional units. However, the in vivo role of each component within the complexes is not known. ATP-dependent chromatin remodeling complexes form several types of protein complexes, which reorganize chromatin structure cooperatively with histone modifiers. Williams syndrome transcription factor (WSTF) was biochemically identified as a major subunit, along with 2 distinct complexes: WINAC, a SWI/SNF-type complex, and WICH, an ISWI-type complex. Here, WSTF(-/-) mice were generated to investigate its function in chromatin remodeling in vivo. Loss of WSTF expression resulted in neonatal lethality, and all WSTF(-/-) neonates and approximately 10% of WSTF(+/-) neonates suffered cardiovascular abnormalities resembling those found in autosomal-dominant Williams syndrome patients. Developmental analysis of WSTF(-/-) embryos revealed that Gja5 gene regulation is aberrant from E9.5, conceivably because of inappropriate chromatin reorganization around the promoter regions where essential cardiac transcription factors are recruited. In vitro analysis in WSTF(-/-) mouse embryonic fibroblast (MEF) cells also showed impaired transactivation functions of cardiac transcription activators on the Gja5 promoter, but the effects were reversed by overexpression of WINAC components. Likewise in WSTF(-/-) MEF cells, recruitment of Snf2h, an ISWI ATPase, to PCNA and cell survival after DNA damage were both defective, but were ameliorated by overexpression of WICH components. Thus, the present study provides evidence that WSTF is shared and is a functionally indispensable subunit of the WICH complex for DNA repair and the WINAC complex for transcriptional control.


Assuntos
Montagem e Desmontagem da Cromatina , Fatores de Transcrição/metabolismo , Animais , Anormalidades Cardiovasculares/genética , Anormalidades Cardiovasculares/metabolismo , Células Cultivadas , Reparo do DNA , Replicação do DNA , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Camundongos , Fatores de Transcrição/genética
11.
J Biol Chem ; 285(24): 18166-76, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20400511

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/química , Histona Desacetilase 1/química , Receptores de Estrogênio/química , Proteínas Repressoras/química , Acetilação , Animais , Diferenciação Celular , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Histona Desacetilase 1/genética , Histonas/química , Humanos , Camundongos , Mutação , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição , Transcrição Gênica , Ativação Transcricional , Receptor ERRalfa Relacionado ao Estrogênio
13.
J Biol Chem ; 285(11): 8084-93, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20054001

RESUMO

The role of aldosterone has been implicated in the metabolic syndrome and cardiovascular diseases. The biological actions of aldosterone are mediated through mineralocorticoid receptor (MR). Nuclear receptor-mediated gene expression is regulated by dynamic and coordinated recruitment of coactivators and corepressors. To identify new coregulators of the MR, full-length MR was used as bait in yeast two-hybrid screening. We isolated NF-YC, one of the subunits of heterotrimeric transcription factor NF-Y. Specific interaction between MR and NF-YC was confirmed by yeast two-hybrid, mammalian two-hybrid, coimmunoprecipitation assays, and fluorescence subcellular imaging. Transient transfection experiments in COS-7 cells demonstrated that NF-YC repressed MR transactivation in a hormone-sensitive manner. Moreover, reduction of NF-YC protein levels by small interfering RNA potentiated hormonal activation of endogenous target genes in stably MR-expressing cells, indicating that NF-YC functions as an agonist-dependent MR corepressor. The corepressor function of NF-YC is selective for MR, because overexpression of NF-YC did not affect transcriptional activity mediated by androgen, progesterone, or glucocorticoid receptors. Chromatin immunoprecipitation experiments showed that endogenous MR and steroid receptor coactivator-1 were recruited to an endogenous ENaC gene promoter in a largely aldosterone-dependent manner, and endogenous NF-YC was sequentially recruited to the same element. Immunohistochemistry showed that endogenous MR and NF-YC were colocalized within the mouse kidney. Although aldosterone induces interaction of the N and C termini of MR, NF-YC inhibited the N/C interaction. These findings indicate that NF-YC functions as a new corepressor of agonist-bound MR via alteration of aldosterone-induced MR conformation.


Assuntos
Aldosterona/metabolismo , Fator de Ligação a CCAAT/metabolismo , Hidrocortisona/metabolismo , Túbulos Renais Coletores/metabolismo , Receptores de Mineralocorticoides/metabolismo , Aldosterona/farmacologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Canais Epiteliais de Sódio/metabolismo , Histona Desacetilases/metabolismo , Humanos , Hidrocortisona/farmacologia , Imuno-Histoquímica , Túbulos Renais Coletores/citologia , Masculino , Camundongos , Regiões Promotoras Genéticas/fisiologia , Estrutura Terciária de Proteína , Receptores Androgênicos/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/agonistas , Receptores de Mineralocorticoides/química , Receptores de Progesterona/metabolismo , Técnicas do Sistema de Duplo-Híbrido
14.
Endocr J ; 58(2): 77-85, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21242649

RESUMO

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.


Assuntos
Montagem e Desmontagem da Cromatina , Síndrome de Williams/genética , Trifosfato de Adenosina/farmacologia , Animais , Anormalidades Cardiovasculares , Montagem e Desmontagem da Cromatina/genética , Montagem e Desmontagem da Cromatina/fisiologia , Cromossomos Humanos Par 7/genética , Epigênese Genética/genética , Epigênese Genética/fisiologia , Transtornos do Crescimento , Humanos , Hipercalcemia , Deficiência Intelectual , Camundongos , Camundongos Knockout , Receptores de Calcitriol/genética , Receptores de Calcitriol/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica/genética , Ativação Transcricional , Síndrome de Williams/diagnóstico
15.
J Biol Chem ; 284(47): 32472-82, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19776015

RESUMO

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.


Assuntos
Cromatina/química , Sistema de Sinalização das MAP Quinases , Fatores de Transcrição/química , Animais , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA , Epigênese Genética , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Fosforilação , Estrutura Terciária de Proteína , Fatores de Transcrição/metabolismo
16.
EMBO Rep ; 9(6): 563-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18451880

RESUMO

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.


Assuntos
Ciclo Celular/fisiologia , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Substâncias Macromoleculares/metabolismo , Linhagem Celular , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Feminino , Humanos , Ativação Transcricional
17.
Mol Cell Biol ; 27(21): 7486-96, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17709391

RESUMO

The orphan nuclear receptor Nurr1 is essential for the development and maintenance of midbrain dopaminergic neurons, the cells that degenerate during Parkinson's disease, by promoting the transcription of genes involved in dopaminergic neurotransmission. Since Nurr1 lacks a classical ligand-binding pocket, it is not clear which factors regulate its activity and how these factors are affected during disease pathogenesis. Since Wnt signaling via beta-catenin promotes the differentiation of Nurr1(+) dopaminergic precursors in vitro, we tested for functional interactions between these systems. We found that beta-catenin and Nurr1 functionally interact at multiple levels. In the absence of beta-catenin, Nurr1 is associated with Lef-1 in corepressor complexes. Beta-catenin binds Nurr1 and disrupts these corepressor complexes, leading to coactivator recruitment and induction of Wnt- and Nurr1-responsive genes. We then identified KCNIP4/calsenilin-like protein as being responsive to concurrent activation by Nurr1 and beta-catenin. Since KCNIP4 interacts with presenilins, the Alzheimer's disease-associated proteins that promote beta-catenin degradation, we tested the possibility that KCNIP4 induction regulates beta-catenin signaling. KCNIP4 induction limited beta-catenin activity in a presenilin-dependent manner, thereby serving as a negative feedback loop; furthermore, Nurr1 inhibition of beta-catenin activity was absent in PS1(-/-) cells or in the presence of small interfering RNAs specific to KCNIP4. These data describe regulatory convergence between Nurr1 and beta-catenin, providing a mechanism by which Nurr1 could be regulated by Wnt signaling.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Wnt/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Núcleo Celular/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , Presenilina-1/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Ratos , Proteínas Repressoras/metabolismo , beta Catenina/metabolismo
20.
Genes Cells ; 13(6): 623-33, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18459961

RESUMO

The status of chromatin during spermatogenesis is dynamically regulated by specific histone codes or stage-specific histone changes. The functional links between such epigenetic regulation and proteins regulating meiosis are largely unknown. In mammals, genes encoded on the Y chromosome are thought to possess male-specific biological functions. While genes located within the azoospermia factor region (AZF) are known to be involved in spermatogenesis, the physiological function of individual genes is not known. SMCY is a gene mapped to the AZF, and in this report, we analyzed the function of SMCY protein during spermatogenesis. Biochemical identification of the proteins with which it interacted showed that SMCY formed a distinct complex with MSH5, a critical meiosis-regulatory protein in the human testicular germ cell line, NEC8. As anticipated, histone H3K4 demethylase activity was detected. Immunohistochemical analysis revealed the co-localization of SMCY with MSH5 at a specific stage of meiotic prophase progression during murine spermatogenesis. Our results suggest that SMCY may have a male-specific function as a histone H3K4 demethylase by recruiting a meiosis-regulatory protein to condensed DNA.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Código das Histonas , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas/metabolismo , Espermatogênese , Animais , Histona Desmetilases , Histona Metiltransferases , Humanos , Masculino , Meiose , Camundongos , Antígenos de Histocompatibilidade Menor , Proteínas Metiltransferases , Testículo/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA