Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
Expert Opin Ther Pat ; 30(1): 1-13, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31825687

RESUMO

Introduction: Peroxisome proliferator-activated receptors (PPARs), PPARα, PPARδ, and PPARγ, play an important role in the regulation of various physiological processes, specifically lipid and energy metabolism and immunity. PPARα agonists (fibrates) and PPARγ agonists (thiazolidinediones) are used for the treatment of hypertriglyceridemia and type 2 diabetes, respectively. PPARδ activation enhances mitochondrial and energy metabolism but PPARδ-acting drugs are not yet available. Many synthetic ligands for PPARs have been developed to expand their therapeutic applications.Areas covered: The authors searched recent patent activity regarding PPAR ligands. Novel PPARα agonists, PPARδ agonists, PPARγ agonists, PPARα/γ dual agonists, and PPARγ antagonists have been claimed for the treatment of metabolic disease and inflammatory disease. Methods for the combination of PPAR ligands with other drugs and expanded application of PPAR agonists for bone and neurological disease have been also claimed.Expert opinion: Novel PPAR ligands and the combination of PPAR ligands with other drugs have been claimed for the treatment of mitochondrial disease, inflammatory/autoimmune disease, neurological disease, and cancer in addition to metabolic diseases including dyslipidemia and type 2 diabetes. Selective therapeutic actions of PPAR ligands should be exploited to avoid adverse effects. More basic studies are needed to elucidate the molecular mechanisms of selective actions.


Assuntos
PPAR alfa/metabolismo , PPAR delta/metabolismo , PPAR gama/metabolismo , Animais , Desenvolvimento de Medicamentos , Humanos , Ligantes , PPAR alfa/agonistas , PPAR alfa/antagonistas & inibidores , PPAR delta/agonistas , PPAR delta/antagonistas & inibidores , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Patentes como Assunto
6.
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
7.
Curr Med Chem ; 24(9): 868-875, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27915991

RESUMO

Vitamin D deficiency and insufficiency are associated with an increased risk of cancer, autoimmune disease, inflammation, infection, cardiovascular disease and metabolic disease, as well as bone and mineral disorders. The vitamin D receptor (VDR), a member of the nuclear receptor superfamily, is a receptor for the active form of vitamin D, 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], and mediates vitamin D regulation of specific target gene expression. The secondary bile acid lithocholic acid, which is produced by intestinal bacteria, is another natural VDR ligand. VDR signaling has been suggested to be involved in reciprocal communication between intestinal cells, including immune and epithelial cells, and intestinal microflora. In addition to epidemiological studies on vitamin D status, genome-wide analyses and cellular and animal experiments have shown that VDR is involved in the prevention of inflammatory bowel disease (IBD) and colorectal cancer (CRC). VDR deletion in mice exaggerates colitis and colon tumorigenesis in experimental models, and treatment of mice with synthetic vitamin D analogues ameliorates pathological changes in these diseases. Several VDR ligands are less active in increasing serum calcium levels, showing higher therapeutic efficiency than the natural hormone 1,25(OH)2D3. VDR plays a role in intestinal homeostasis and in protection against IBD and CRC. The development of VDR ligands with reduced or no calcemic activity will be necessary to expand clinical application of VDRtargeting therapy.


Assuntos
Neoplasias Colorretais/prevenção & controle , Doenças Inflamatórias Intestinais/prevenção & controle , Receptores de Calcitriol/metabolismo , Animais , Humanos , Ligantes
8.
Immunobiology ; 221(2): 188-92, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26386981

RESUMO

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS) caused by the infiltration of TH1 and TH17 cells into the CNS. Ribosomal S6 kinase 2 (RSK2; RPS6KA3) regulates TH17 differentiation by attenuating RORγt transcriptional activities and IL-17A production. The pan-RSK inhibitor BI-D1870 also inhibits TH17 differentiation, but the effect of BI-D1870 in vivo remains unclear. Here, we generated mice with experimental autoimmune encephalomyelitis (EAE) and treated them with BI-D1870. BI-D1870 administration protected mice from EAE by reducing the infiltration of TH1 and TH17 cells into the CNS and decreasing mRNA levels of Ccr6 in TH17 cells. These results suggest that RSK inhibition is a promising strategy for the treatment of MS.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Pteridinas/farmacologia , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Movimento Celular/efeitos dos fármacos , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Receptores CCR6/antagonistas & inibidores , Receptores CCR6/genética , Receptores CCR6/imunologia , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/imunologia , Transdução de Sinais , Células Th1/imunologia , Células Th1/patologia , Células Th17/imunologia , Células Th17/patologia
9.
Expert Opin Ther Pat ; 25(12): 1373-83, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26402335

RESUMO

INTRODUCTION: The vitamin D receptor (VDR) is a promising drug target in the treatment of cancer, autoimmune disease, inflammation, infection and cardiovascular disease, as well as bone and mineral disorders. Although many VDR ligands have been developed and shown to activate VDR in vitro and in vivo, including vitamin D derivatives and non-secosteroidal compounds, a principal adverse effect of hypercalcemia has limited their clinical application. AREAS COVERED: We summarize recent patent activity regarding VDR ligands, including vitamin D derivatives, non-secosteroidal compounds and tissue-selective prodrugs, alongside their therapeutic applications. The potential for use of VDR ligands in the treatment of hepatic fibrosis, pancreatic fibrosis and neuronal disease is also reviewed. EXPERT OPINION: Several VDR ligands have been shown to have increased therapeutic efficiency in experimental models of cancer, inflammation and cardiovascular disease, and to exhibit function-selective and/or tissue-selective activity. The underlying molecular and pharmacological mechanisms remain to be elucidated. Further studies, both basic and applied, should make successful VDR-targeting therapy possible.


Assuntos
Desenho de Fármacos , Receptores de Calcitriol/efeitos dos fármacos , Vitamina D/administração & dosagem , Animais , Humanos , Hipercalcemia/induzido quimicamente , Ligantes , Terapia de Alvo Molecular , Patentes como Assunto , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Vitamina D/farmacologia , Vitaminas/efeitos adversos , Vitaminas/farmacologia , Vitaminas/uso terapêutico
11.
Mol Cell Biol ; 35(2): 344-55, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368387

RESUMO

The Dgcr14/Es2 gene is located in a chromosomal region the loss of which has been associated with DiGeorge syndrome, a cause of immunodeficiency, heart defects, and skeletal abnormalities. However, the role of DGCR14 protein remains to be elucidated. Here, I found that DGCR14 protein acts as a coactivator of RORγt in TH17 cells. Biochemical purification of the RORγ coregulator complex allowed me to identify the associated DGCR14 protein by matrix-assisted laser desorption ionization-time of flight mass spectrometry. Overexpression of Dgcr14 mRNA enhanced RORγt-mediated transcriptional activity and facilitated TH17 cell differentiation. Furthermore, knockdown of Dgcr14 reduced Il17a mRNA expression. I also found that DGCR14 associated with ribosomal S6 kinase 2 (RSK2, also called RpS6ka3) and BAZ1B, both of which were recruited to the Il17a promoter during TH17 cell differentiation. Knockdown of Baz1b or RpS6ka3 also reduced Il17a mRNA expression, and Baz1b knockdown increased transcriptional suppressive histone marks (histone H3K9me3) on the Il17a promoter. My findings showed the roles of DGCR14, RSK2, and BAZ1B in the transcriptional regulation of Il17a mRNA during TH17 cell differentiation.


Assuntos
Interleucina-17/genética , Proteínas Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica/genética , Histonas/metabolismo , Humanos , Camundongos , RNA Mensageiro/metabolismo , Transdução de Sinais/genética
12.
Expert Opin Ther Pat ; 25(2): 175-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25416646

RESUMO

INTRODUCTION: PPARγ regulates glucose and lipid metabolism, immunity, and cellular growth and differentiation. Thiazolidinediones (TZDs) are synthetic PPARγ ligands that are used in the treatment of type 2 diabetes. However, TZDs can cause adverse effects, such as increased risks of heart failure, bone fractures and bladder cancer. PPARγ has a large ligand-binding pocket, which makes it possible to develop a variety of PPARγ ligands, leading to patents on their therapeutic applications. AREAS COVERED: We summarize recent patent activity regarding PPARγ ligands and their therapeutic applications from 2008. Pharmacologic methods to increase PPARγ expression and to decrease PPARγ adverse effects by combining PPARγ ligands with other drugs are also reviewed. EXPERT OPINION: In addition to novel PPARγ ligands that exhibit selective therapeutic activity without adverse effects, such as bone loss, fluid retention or weight gain, methods for the combination of PPARγ ligands and other compounds have been claimed to decrease adverse effects and/or to enhance targeted effects. Combination therapy is useful but has the potential risk of unexpected adverse effects. Patent applications for expanding clinical application of PPARγ ligands to non-metabolic diseases, such as neurological and inflammatory diseases, and to skin whitening have been filed, and future studies are needed to elucidate the underlying mechanisms.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , PPAR gama/efeitos dos fármacos , Animais , Desenho de Fármacos , Humanos , Hipoglicemiantes/efeitos adversos , Ligantes , Metabolismo dos Lipídeos , PPAR gama/metabolismo , Patentes como Assunto , Tiazolidinedionas/efeitos adversos , Tiazolidinedionas/farmacologia
14.
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
15.
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
16.
Nat Med ; 18(3): 405-12, 2012 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-22344299

RESUMO

The signaling molecule Wnt regulates bone homeostasis through ß-catenin-dependent canonical and ß-catenin-independent noncanonical pathways. Impairment of canonical Wnt signaling causes bone loss in arthritis and osteoporosis; however, it is unclear how noncanonical Wnt signaling regulates bone resorption. Wnt5a activates noncanonical Wnt signaling through receptor tyrosine kinase-like orphan receptor (Ror) proteins. We showed that Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhanced osteoclastogenesis. Osteoblast-lineage cells expressed Wnt5a, whereas osteoclast precursors expressed Ror2. Mice deficient in either Wnt5a or Ror2, and those with either osteoclast precursor-specific Ror2 deficiency or osteoblast-lineage cell-specific Wnt5a deficiency showed impaired osteoclastogenesis. Wnt5a-Ror2 signals enhanced receptor activator of nuclear factor-κB (RANK) expression in osteoclast precursors by activating JNK and recruiting c-Jun on the promoter of the gene encoding RANK, thereby enhancing RANK ligand (RANKL)-induced osteoclastogenesis. A soluble form of Ror2 acted as a decoy receptor of Wnt5a and abrogated bone destruction in mouse arthritis models. Our results suggest that the Wnt5a-Ror2 pathway is crucial for osteoclastogenesis in physiological and pathological environments and represents a therapeutic target for bone diseases, including arthritis.


Assuntos
Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Artrite/metabolismo , Doenças Ósseas/metabolismo , Linhagem da Célula , Regulação da Expressão Gênica , MAP Quinase Quinase 4/metabolismo , Camundongos , Osteoblastos/citologia , Osteoclastos/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/deficiência , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Crânio/citologia , Crânio/crescimento & desenvolvimento , Proteínas Wnt/deficiência , Proteínas Wnt/genética , Proteína Wnt-5a , Microtomografia por Raio-X
17.
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
18.
Biosci Biotechnol Biochem ; 74(9): 1956-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20834143

RESUMO

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.


Assuntos
Anexinas/análise , Apoferritinas/análise , Reabsorção Óssea/etiologia , Proteínas de Transporte/análise , Membrana Nuclear/química , Osteoclastos/química , Animais , Linhagem Celular , Células Cultivadas , Eletroforese em Gel Bidimensional , Redes Reguladoras de Genes , Macrófagos , Camundongos
19.
Ann N Y Acad Sci ; 1192: 201-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20392237

RESUMO

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.


Assuntos
Proteínas Correpressoras/isolamento & purificação , Receptor alfa de Estrogênio/metabolismo , Osteoclastos/metabolismo , Transativadores/isolamento & purificação , Animais , Clonagem Molecular , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Receptor alfa de Estrogênio/isolamento & purificação , Feminino , Humanos , Camundongos , Modelos Biológicos , Osteoclastos/química , Transativadores/genética , Transativadores/metabolismo , Ativação Transcricional/genética
20.
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