Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 298(9): 102342, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35933014

RESUMO

Ess2, also known as Dgcr14, is a transcriptional co-regulator of CD4+ T cells. Ess2 is located in a chromosomal region, the loss of which has been associated with 22q11.2 deletion syndrome (22q11DS), which causes heart defects, skeletal abnormalities, and immunodeficiency. However, the specific association of Ess2 with 22q11DS remains unclear. To elucidate the role of Ess2 in T-cell development, we generated Ess2 floxed (Ess2fl/fl) and CD4+ T cell-specific Ess2 KO (Ess2ΔCD4/ΔCD4) mice using the Cre/loxP system. Interestingly, Ess2ΔCD4/ΔCD4 mice exhibited reduced naïve T-cell numbers in the spleen, while the number of thymocytes (CD4-CD8-, CD4+CD8+, CD4+CD8-, and CD4-CD8+) in the thymus remained unchanged. Furthermore, Ess2ΔCD4/ΔCD4 mice had decreased NKT cells and increased γδT cells in the thymus and spleen. A genome-wide expression analysis using RNA-seq revealed that Ess2 deletion alters the expression of many genes in CD4 single-positive thymocytes, including genes related to the immune system and Myc target genes. In addition, Ess2 enhanced the transcriptional activity of c-Myc. Some genes identified as Ess2 targets in mice show expressional correlation with ESS2 in human immune cells. Moreover, Ess2ΔCD4/ΔCD4 naïve CD4+ T cells did not maintain survival in response to IL-7. Our results suggest that Ess2 plays a critical role in post-thymic T-cell survival through the Myc and IL-7 signaling pathways.


Assuntos
Linfócitos T CD4-Positivos , Interleucina-7 , Proteínas Nucleares , Proteínas Proto-Oncogênicas c-myc , Transcrição Gênica , Animais , Humanos , Camundongos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/genética , Sobrevivência Celular , Interleucina-7/metabolismo , Camundongos Knockout , Células T Matadoras Naturais/imunologia , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Timo/citologia , Timo/imunologia
2.
Cancer Sci ; 113(2): 587-596, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34807483

RESUMO

Checkpoint kinase 1 (CHK1) plays a key role in genome surveillance and integrity throughout the cell cycle. Selective inhibitors of CHK1 (CHK1i) are undergoing clinical evaluation for various human malignancies, including neuroblastoma. In this study, one CHK1i-sensitive neuroblastoma cell line, CHP134, was investigated, which characteristically carries MYCN amplification and a chromosome deletion within the 10q region. Among several cancer-related genes in the chromosome 10q region, mRNA expression of fibroblast growth factor receptor 2 (FGFR2) was altered in CHP134 cells and associated with an unfavorable prognosis of patients with neuroblastoma. Induced expression of FGFR2 in CHP134 cells reactivated downstream MEK/ERK signaling and resulted in cells resistant to CHK1i-mediated cell growth inhibition. Consistently, the MEK1/2 inhibitor, trametinib, potentiated CHK1 inhibitor-mediated cell death in these cells. These results suggested that FGFR2 loss might be prone to highly effective CHK1i treatment. In conclusion, extreme cellular dependency of ERK activation may imply a possible application for the MEK1/2 inhibitor, either as a single inhibitor or in combination with CHK1i in MYCN-amplified neuroblastomas.


Assuntos
Apoptose/efeitos dos fármacos , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Proteína Proto-Oncogênica N-Myc/genética , Inibidores de Proteínas Quinases/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Amplificação de Genes , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Prognóstico , Piridonas/farmacologia , Pirimidinonas/farmacologia , RNA Mensageiro/genética
3.
Physiol Rev ; 93(2): 481-523, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23589826

RESUMO

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.


Assuntos
Desenvolvimento Ósseo/genética , Osso e Ossos/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , Animais , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/citologia , Osso e Ossos/fisiopatologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Ligantes
4.
Biochem Biophys Res Commun ; 497(2): 597-604, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-29454968

RESUMO

Transcription and pre-mRNA splicing are complex, coupled processes that involve transcriptional co-regulators. Ess2 (also termed Dgcr14) is a nuclear protein that enhances the transcriptional activity of retinoic acid receptor-related orphan receptor gamma/gamma-t (Rorγ/γt). Ess2 is also a component of the spliceosomal C complex (containing U2, U5 and U6 snRNAs). However, the domains in Ess2 that function in splicing and transcription have not been identified. To elucidate the roles of Ess2 in splicing and transcription, we performed RNA immunoprecipitation (RIP) assays to detect Ess2-interacting snRNAs. We found that Ess2 associated with U6 snRNA as well as U1 and U4 snRNAs. Experiments using Ess2 deletion mutants showed that a C-terminus deletion mutant of Ess2 (1-399 a. a.) lost its ability to associate with snRNAs, whereas the N-terminus domain of Ess2 (1-200 a. a.) associated with Rorγ/γt, but not with snRNAs. Interestingly, experiments using anti-ROR common antibody showed that Rors also associated with U4 and U6 snRNAs. Ess2 knockdown in a T cell hybridoma (68-41 cells) abrogated the interaction between spliceosomes and Rors. An Ess2-dependent association was also found between an lncRNA (Rmrp) and Rors. We thus propose that Ess2 associates with both transcriptional factors and spliceosomal complexes and modulates splicing reactions coupled with transcription factors.


Assuntos
Proteínas Nucleares/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , RNA Nuclear Pequeno/metabolismo , Spliceossomos/metabolismo , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Mutação , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Domínios e Motivos de Interação entre Proteínas , Splicing de RNA , RNA Nuclear Pequeno/genética , Spliceossomos/genética , Ativação Transcricional
5.
Int J Mol Sci ; 19(7)2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29958417

RESUMO

Farnesoid X receptor (FXR) is a receptor for bile acids and plays an important role in the regulation of bile acid metabolism in the liver. Although FXR has been shown to affect hepatocarcinogenesis through both direct and indirect mechanisms, potential roles of FXR in epithelial­mesenchymal transition (EMT) in hepatocellular carcinoma (HCC) remain unclear. We examined the effect of several FXR ligands on EMT-related morphological changes in HCC cell lines, such as HuH-7 and Hep3B cells. FXR agonists (chenodeoxycholic acid, GW4064, and obeticholic acid)­but not an antagonist (guggulsterone)—induced actin polymerization and expression of N-cadherin and phosphorylated focal adhesion kinase, although they were less effective than transforming growth factor ß (TGF-ß). FXR agonist treatment enhanced TGF-ß-induced EMT morphologic changes and FXR antagonist inhibited the effect of TGF-ß. Thus, FXR activation enhances EMT in HCC and FXR antagonists may be EMT-suppressing drug candidates.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Receptores Citoplasmáticos e Nucleares/genética , Fator de Crescimento Transformador beta1/genética , Ácidos e Sais Biliares/metabolismo , Caderinas/genética , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacologia , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Isoxazóis/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores
6.
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
7.
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
8.
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
9.
Proc Natl Acad Sci U S A ; 108(12): 4938-43, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21383160

RESUMO

Prostate cancer development is associated with hyperactive androgen signaling. However, the molecular link between androgen receptor (AR) function and humoral factors remains elusive. A prostate cancer mouse model was generated by selectively mutating the AR threonine 877 into alanine in prostatic epithelial cells through Cre-ERT2-mediated targeted somatic mutagenesis. Such AR point mutant mice (ARpe-T877A/Y) developed hypertrophic prostates with responses to both an androgen antagonist and estrogen, although no prostatic tumor was seen. In prostate cancer model transgenic mice, the onset of prostatic tumorigenesis as well as tumor growth was significantly potentiated by introduction of the AR T877A mutation into the prostate. Genetic screening of mice identified Wnt-5a as an activator. Enhanced Wnt-5a expression was detected in the malignant prostate tumors of patients, whereas in benign prostatic hyperplasia such aberrant up-regulation was not obvious. These findings suggest that a noncanonical Wnt signal stimulates development of prostatic tumors with AR hyperfunction.


Assuntos
Androgênios/metabolismo , Neoplasias Experimentais/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Substituição de Aminoácidos , Androgênios/genética , Animais , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/genética , Mutação Puntual , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Proteínas Wnt/genética
10.
Int Immunol ; 24(4): 253-65, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22331441

RESUMO

Although transforming growth factor (TGF)-ß1 is a well-known immunosuppressive cytokine, little is known about the role of its downstream transcription factors, Smad2 and Smad3, in the suppression of macrophage activation. Previous studies have demonstrated that Smad3 is critical for the suppression of LPS-mediated inducible nitric oxide (NO) synthase (iNOS) induction, although the role of Smad2 remains to be investigated. In this study, we found that iNOS induction was enhanced in Smad2-deficient bone marrow-derived macrophages (BMDMs) and peritoneal macrophages in vitro and tumor-associated macrophages in vivo, compared with wild-type (WT) macrophages. However, TGF-ß1 still suppressed iNOS induction in Smad2-deficient macrophages. In Smad2/3 double knockout (KO) (Smad2/3 DKO) BMDMs, LPS-mediated NO/iNOS induction was more strongly elevated than in Smad2 or Smad3 single KO BMDMs, and its suppression by exogenous TGF-ß1 was severely impaired. These data suggest that Smad2 and Smad3 redundantly regulate iNOS induction. Similarly, the production of IL-6 and TNFα, but not IL-10 was augmented in Smad2/3 DKO BMDMs, suggesting that Smad2 and Smad3 also redundantly suppressed some cytokines production. In Smad2/3 DKO macrophages, TLR3- as well as TLR4-mediated IRF3 activation and IFN-ß production were strongly augmented, which resulted in hyper STAT1 phosphorylation. Furthermore, IFN-ß- and IFN-γ-induced iNOS induction in the absence of TLR signaling and STAT1 transcriptional activity were augmented in Smad2/3 DKO BMDMs. These results suggest that Smad2 and Smad3 negatively regulate iNOS induction in macrophages by suppressing multiple steps in the IRF3-IFN-ß-STAT1 pathway.


Assuntos
Macrófagos/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Animais , Células da Medula Óssea/imunologia , Células Cultivadas , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/biossíntese , Interferon beta/metabolismo , Interferon gama/metabolismo , Interleucina-10 , Interleucina-6/biossíntese , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Proteína Smad2/deficiência , Proteína Smad2/genética , Proteína Smad3/deficiência , Proteína Smad3/genética , Receptor 3 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Transcrição Gênica , Fator de Crescimento Transformador beta1/imunologia , Fator de Crescimento Transformador beta1/metabolismo
11.
Nature ; 446(7135): 562-6, 2007 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-17392787

RESUMO

Fat-soluble ligands, including sex steroid hormones and environmental toxins, activate ligand-dependent DNA-sequence-specific transcriptional factors that transduce signals through target-gene-selective transcriptional regulation. However, the mechanisms of cellular perception of fat-soluble ligand signals through other target-selective systems remain unclear. The ubiquitin-proteasome system regulates selective protein degradation, in which the E3 ubiquitin ligases determine target specificity. Here we characterize a fat-soluble ligand-dependent ubiquitin ligase complex in human cell lines, in which dioxin receptor (AhR) is integrated as a component of a novel cullin 4B ubiquitin ligase complex, CUL4B(AhR). Complex assembly and ubiquitin ligase activity of CUL4B(AhR) in vitro and in vivo are dependent on the AhR ligand. In the CUL4B(AhR) complex, ligand-activated AhR acts as a substrate-specific adaptor component that targets sex steroid receptors for degradation. Thus, our findings uncover a function for AhR as an atypical component of the ubiquitin ligase complex and demonstrate a non-genomic signalling pathway in which fat-soluble ligands regulate target-protein-selective degradation through a ubiquitin ligase complex.


Assuntos
Proteínas Culina/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Linhagem Celular , Proteínas Culina/química , Proteínas Culina/genética , Receptor alfa de Estrogênio/metabolismo , Humanos , Ligantes , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Ligação Proteica , Receptores de Hidrocarboneto Arílico/genética , Especificidade por Substrato , Ativação Transcricional
13.
Asian J Androl ; 25(2): 158-165, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36695247

RESUMO

Prostate cancer is one of the most common diseases in men worldwide. Surgery, radiation therapy, and hormonal therapy are effective treatments for early-stage prostate cancer. However, the development of castration-resistant prostate cancer has increased the mortality rate of prostate cancer. To develop novel drugs for castration-resistant prostate cancer, the molecular mechanisms of prostate cancer progression must be elucidated. Among the signaling pathways regulating prostate cancer development, recent studies have revealed the importance of noncanonical wingless-type MMTV integration site family (WNT) signaling pathways, mainly that involving WNT5A, in prostate cancer progression and metastasis; however, its role remains controversial. Moreover, chromatin remodelers such as the switch/sucrose nonfermentable (SWI/SNF) complex and chromodomain helicase DNA-binding proteins 1 also play important roles in prostate cancer progression through genome-wide gene expression changes. Here, we review the roles of noncanonical WNT signaling pathways, chromatin remodelers, and epigenetic enzymes in the development and progression of prostate cancer.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Via de Sinalização Wnt , Masculino , Humanos , Cromatina , Montagem e Desmontagem da Cromatina
14.
Sci Rep ; 13(1): 12355, 2023 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-37524814

RESUMO

Molecular targeted therapy using poly (ADP-ribose) polymerase inhibitors has improved survival in patients with castration-resistant prostate cancer (CRPC). However, this approach is only effective in patients with specific genetic mutations, and additional drug discovery targeting epigenetic modulators is required. Here, we evaluated the involvement of the transcriptional coregulator ESS2 in prostate cancer. ESS2-knockdown PC3 cells dramatically inhibited proliferation in tumor xenografts in nude mice. Microarray analysis revealed that ESS2 regulated mRNA levels of chromodomain helicase DNA binding protein 1 (CHD1)-related genes and other cancer-related genes, such as PPAR-γ, WNT5A, and TGF-ß, in prostate cancer. ESS2 knockdown reduced nuclear factor (NF)-κB/CHD1 recruitment and histone H3K36me3 levels on the promoters of target genes (TNF and CCL2). In addition, we found that the transcriptional activities of NF-κB, NFAT and SMAD2/3 were enhanced by ESS2. Tamoxifen-inducible Ess2-knockout mice showed delayed prostate development with hypoplasia and disruption of luminal cells in the ventral prostate. Overall, these findings identified ESS2 acts as a transcriptional coregulator in prostate cancer and ESS2 can be novel epigenetic therapeutic target for CRPC.


Assuntos
Próstata , Neoplasias de Próstata Resistentes à Castração , Masculino , Animais , Camundongos , Humanos , Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Camundongos Nus , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , NF-kappa B/metabolismo , Processos Neoplásicos , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Proliferação de Células
15.
J Biol Chem ; 286(41): 35456-35465, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21862595

RESUMO

Suppression of IL-2 ßproduction from T cells is an important process for the immune regulation by TGF-ß. However, the mechanism by which this suppression occurs remains to be established. Here, we demonstrate that Smad2 and Smad3, two major TGF-ß-downstream transcription factors, are redundantly essential for TGF-ß-mediated suppression of IL-2 production in CD4(+) T cells using Smad2- and Smad3-deficient T cells. Both Smad2 and Smad3 were recruited into the proximal region of the IL-2 promoter in response to TGF-ß. We then investigated the histone methylation status of the IL-2 promoter. Although both histone H3 lysine 9 (H3K9) and H3K27 trimethylation have been implicated in gene silencing, only H3K9 trimethylation was increased in the proximal region of the IL-2 promoter in a Smad2/3-dependent manner, whereas H3K27 trimethylation was not. The H3K9 methyltransferases Setdb1 and Suv39h1 bound to Smad3 and suppressed IL-2 promoter activity in collaboration with Smad3. Overexpression of Suv39h1 in 68-41 T cells strongly inhibited IL-2 production in response to T cell receptor stimulation irrespective of the presence or absence of TGF-ß, whereas Setdb1 overexpression only slightly suppressed IL-2 production. Silencing of Suv39h1 by shRNA reverted the suppressive effect of TGF-ß on IL-2 production. Furthermore, TGF-ß induced Suv39h1 recruitment to the proximal region of the IL-2 promoter in wild type primary T cells; however, this was not observed in Smad2(-/-)Smad3(+/-) T cells. Thus, we propose that Smads recruit H3K9 methyltransferases Suv39h1 to the IL-2 promoter, thereby inducing suppressive histone methylation and inhibiting T cell receptor-mediated IL-2 transcription.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Interleucina-2/biossíntese , Metiltransferases/metabolismo , Regiões Promotoras Genéticas/fisiologia , Proteínas Repressoras/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Transcrição Gênica/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Inativação Gênica , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/imunologia , Histonas/genética , Histonas/imunologia , Histonas/metabolismo , Interleucina-2/genética , Interleucina-2/imunologia , Metilação , Metiltransferases/genética , Metiltransferases/imunologia , Camundongos , Camundongos Knockout , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Proteína Smad2/genética , Proteína Smad2/imunologia , Proteína Smad3/genética , Proteína Smad3/imunologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia
16.
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
18.
Clin Calcium ; 22(5): 629-36, 2012 May.
Artigo em Japonês | MEDLINE | ID: mdl-22549186

RESUMO

To maintain a balance between the differentiation of adipocytes and osteoblasts is one of significant issues related to osteoporosis and metabolic syndrome. Both adipocytes and osteoblasts are derived from mesenchymal stem cells and recent studies have shown that osteoblast or adipocyte differentiation regulators reciprocally control each cell differentiations. In particular, PPAR-gamma plays a pivotal role for adipocyte differentiation and directly controls the differentiation of osteoblast and osteoclast. In this report, I summarize recent researches mainly focusing on the transcriptional co-regulators and extracellular signals regulating the transactivation function of PPAR-gamma.


Assuntos
Adipócitos/citologia , Diferenciação Celular/fisiologia , Osteoblastos/citologia , PPAR gama/metabolismo , Transdução de Sinais , Adipócitos/metabolismo , Animais , Diferenciação Celular/genética , Humanos , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , PPAR gama/genética
19.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA