Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Pathol ; 257(3): 352-366, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35297529

RESUMO

Muscular dystrophies are genetic diseases characterized by chronic inflammation and fibrosis. Macrophages are immune cells that sustain muscle regeneration upon acute injury but seem deleterious in the context of chronic muscle injury such as in muscular dystrophies. Here, we observed that the number of macrophages expressing the transcription factor Nfix increases in two distinct mouse models of muscular dystrophies. We showed that the deletion of Nfix in macrophages in dystrophic mice delays the establishment of fibrosis and muscle wasting, and increases grasp force. Macrophages lacking Nfix expressed more TNFα and less TGFß1, thus promoting apoptosis of fibro-adipogenic progenitors. Moreover, pharmacological treatment of dystrophic mice with a ROCK inhibitor accelerated fibrosis through the increase of Nfix expression by macrophages. Thus, we have identified Nfix as a macrophage profibrotic factor in muscular dystrophies, whose inhibition could be a therapeutic route to reduce severity of the dystrophic disease. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Macrófagos , Distrofias Musculares , Fatores de Transcrição NFI , Animais , Fibrose , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Fatores de Transcrição NFI/deficiência , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo
2.
Cell Tissue Res ; 374(3): 531-540, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30091046

RESUMO

Enamel makes up the outermost layer of the crown and its hardness protects other dental tissues from various stimuli. Enamel cannot be regenerated once damaged because ameloblasts are lost during the tooth eruption. Since the ameloblast differentiation mechanism is still unknown, further research is essential for developing treatments for defective or damaged enamel. Previously, we have reported that osteoblast differentiation and bone formation were regulated through the runt-related transcription factor 2 (Runx2)-nuclear factor 1-C (Nfic)-osterix (Osx) pathway where Nfic directly controls Osx expression. This pathway regulates odontoblast differentiation and dentin formation as well. The aim of this study was to investigate if the same pathway is applicable for ameloblast differentiation. Structural enamel defects with disorganized ameloblasts and decreased proliferation activity of the cervical loop were observed in Nfic-/- mice incisors. Expression of the ameloblast differentiation markers was also downregulated significantly in Nfic-/- mice. Real-time PCR analyses suggested that Runx2, Nfic, and Osx regulate the expression of ameloblast differentiation markers, where Runx2 is upstream of Nfic, and Nfic controls Osx expression. Therefore, we suggest the Runx2-Nfic-Osx pathway as one of the key factors that regulate ameloblast differentiation.


Assuntos
Ameloblastos/citologia , Ameloblastos/metabolismo , Diferenciação Celular , Esmalte Dentário/metabolismo , Fatores de Transcrição NFI/metabolismo , Transdução de Sinais , Fator de Transcrição Sp7/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Linhagem da Célula , Proliferação de Células , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Esmalte Dentário/ultraestrutura , Camundongos , Fatores de Transcrição NFI/deficiência , Dente/metabolismo , Dente/ultraestrutura , Microtomografia por Raio-X
3.
Biol Chem ; 397(11): 1173-1185, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27276529

RESUMO

The protein inhibitor of activated STAT1 (PIAS1) plays important roles in regulating virus-induced chronic hepatitis, but the interaction between hepatitis B virus (HBV) and hPIAS1 is not clear. Our aim was to verify if HBV encoding proteins enhance the transcription of hPIAS1 and which cis-elements and transcription factors were involved in the mechanism. In order to do, so a series of molecular biological methods, along with functional and histological studies, were performed. We found that the HBV surface protein (HBs) enhanced hPIAS1 transcription through the activities of TAL1, E47, myogenin (MYOG), and NFI, dependent on the activation of p38MAPK and ERK signaling pathways in vitro, which might contribute to the ineffectiveness of treatment in CHB patients. Furthermore, liver samples from patients with high HBsAg levels and HBV DNA displayed increased hPIAS1 expression and high levels of TAL1, E47, MYOG, and NFI, compared to those patients with low HBsAg levels and HBV DNA, and healthy controls. These findings suggest that the HBs protein-induced hPIAS1 transcription requires TAL1, E47, MYOG, NFI, and MAPK signal pathways. It provides new potential targets for antiviral therapeutic strategies for controlling HBV-associated diseases.


Assuntos
Vírus da Hepatite B/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Nucleares/metabolismo , Proteínas Inibidoras de STAT Ativados/genética , Transcrição Gênica , Proteínas do Envelope Viral/metabolismo , Adulto , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células CHO , Cricetinae , Cricetulus , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células Hep G2 , Antígenos de Superfície da Hepatite B/metabolismo , Vírus da Hepatite B/fisiologia , Humanos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miogenina/genética , Miogenina/metabolismo , Fatores de Transcrição NFI/deficiência , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fosforilação , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fator 3 de Transcrição/deficiência , Fator 3 de Transcrição/genética , Fator 3 de Transcrição/metabolismo
4.
Blood ; 122(17): 2987-96, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-24041575

RESUMO

Hematopoietic stem cells are both necessary and sufficient to sustain the complete blood system of vertebrates. Here we show that Nfix, a member of the nuclear factor I (Nfi) family of transcription factors, is highly expressed by hematopoietic stem and progenitor cells (HSPCs) of murine adult bone marrow. Although short hairpin RNA-mediated knockdown of Nfix expression in Lineage(-)Sca-1(+)c-Kit(+) HSPCs had no effect on in vitro cell growth or viability, Nfix-depleted HSPCs displayed a significant loss of colony-forming potential, as well as short- and long-term in vivo hematopoietic repopulating activity. Analysis of recipient mice at 4 to 20 days posttransplant revealed that Nfix-depleted HSPCs are established in the bone marrow, but fail to persist due to increased apoptotic cell death. Gene expression profiling of Nfix-depleted HSPCs reveals that loss of Nfix expression in HSPCs is concomitant with a decrease in the expression of multiple genes known to be important for HSPCs survival, such as Erg, Mecom, and Mpl. These data reveal that Nfix is a novel regulator of HSPCs survival posttransplantation and establish a role for Nfi genes in the regulation of this cellular compartment.


Assuntos
Células-Tronco Adultas/metabolismo , Células da Medula Óssea/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Fatores de Transcrição NFI/genética , Células-Tronco Adultas/citologia , Animais , Antígenos Ly/genética , Antígenos Ly/metabolismo , Apoptose , Células da Medula Óssea/citologia , Sobrevivência Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Fatores de Transcrição NFI/deficiência , Fatores de Transcrição NFI/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Trombopoetina/genética , Receptores de Trombopoetina/metabolismo , Transdução de Sinais , Fatores de Transcrição , Regulador Transcricional ERG
5.
Nature ; 495(7439): 98-102, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23389444

RESUMO

Adult stem cells reside in specialized niches where they receive environmental cues to maintain tissue homeostasis. In mammals, the stem cell niche within hair follicles is home to epithelial hair follicle stem cells and melanocyte stem cells, which sustain cyclical bouts of hair regeneration and pigmentation. To generate pigmented hairs, synchrony is achieved such that upon initiation of a new hair cycle, stem cells of each type activate lineage commitment. Dissecting the inter-stem-cell crosstalk governing this intricate coordination has been difficult, because mutations affecting one lineage often affect the other. Here we identify transcription factor NFIB as an unanticipated coordinator of stem cell behaviour. Hair follicle stem-cell-specific conditional targeting of Nfib in mice uncouples stem cell synchrony. Remarkably, this happens not by perturbing hair cycle and follicle architecture, but rather by promoting melanocyte stem cell proliferation and differentiation. The early production of melanin is restricted to melanocyte stem cells at the niche base. Melanocyte stem cells more distant from the dermal papilla are unscathed, thereby preventing hair greying typical of melanocyte stem cell differentiation mutants. Furthermore, we pinpoint KIT-ligand as a dermal papilla signal promoting melanocyte stem cell differentiation. Additionally, through chromatin-immunoprecipitation with high-throughput-sequencing and transcriptional profiling, we identify endothelin 2 (Edn2) as an NFIB target aberrantly activated in NFIB-deficient hair follicle stem cells. Ectopically induced Edn2 recapitulates NFIB-deficient phenotypes in wild-type mice. Conversely, endothelin receptor antagonists and/or KIT blocking antibodies prevent precocious melanocyte stem cell differentiation in the NFIB-deficient niche. Our findings reveal how melanocyte and hair follicle stem cell behaviours maintain reliance upon cooperative factors within the niche, and how this can be uncoupled in injury, stress and disease states.


Assuntos
Folículo Piloso/citologia , Melanócitos/citologia , Fatores de Transcrição NFI/metabolismo , Nicho de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Imunoprecipitação da Cromatina , Endotelina-2/genética , Endotelina-2/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Cabelo/citologia , Cabelo/crescimento & desenvolvimento , Cor de Cabelo , Folículo Piloso/metabolismo , Melanócitos/metabolismo , Camundongos , Fatores de Transcrição NFI/deficiência , Fatores de Transcrição NFI/genética , Análise de Sequência , Fator de Células-Tronco/metabolismo
6.
J Neurosci ; 30(27): 9127-39, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20610746

RESUMO

The balance between self-renewal and differentiation of neural progenitor cells is an absolute requirement for the correct formation of the nervous system. Much is known about both the pathways involved in progenitor cell self-renewal, such as Notch signaling, and the expression of genes that initiate progenitor differentiation. However, whether these fundamental processes are mechanistically linked, and specifically how repression of progenitor self-renewal pathways occurs, is poorly understood. Nuclear factor I A (Nfia), a gene known to regulate spinal cord and neocortical development, has recently been implicated as acting downstream of Notch to initiate the expression of astrocyte-specific genes within the cortex. Here we demonstrate that, in addition to activating the expression of astrocyte-specific genes, Nfia also downregulates the activity of the Notch signaling pathway via repression of the key Notch effector Hes1. These data provide a significant conceptual advance in our understanding of neural progenitor differentiation, revealing that a single transcription factor can control both the activation of differentiation genes and the repression of the self-renewal genes, thereby acting as a pivotal regulator of the balance between progenitor and differentiated cell states.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição NFI/fisiologia , Células-Tronco/fisiologia , Telencéfalo/citologia , Fatores Etários , Análise de Variância , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/embriologia , Imunoprecipitação da Cromatina/métodos , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries/métodos , Mutação/genética , Fatores de Transcrição NFI/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fator 6 de Transcrição de Octâmero/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas/fisiologia , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Telencéfalo/embriologia , Fatores de Transcrição HES-1 , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
7.
Mol Cell Biol ; 29(22): 6006-17, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19752192

RESUMO

Transforming growth factor beta (TGF-beta) and platelet-derived growth factor A (PDGFAlpha) play a central role in tissue morphogenesis and repair, but their interplay remain poorly understood. The nuclear factor I C (NFI-C) transcription factor has been implicated in TGF-beta signaling, extracellular matrix deposition, and skin appendage pathologies, but a potential role in skin morphogenesis or healing had not been assessed. To evaluate this possibility, we performed a global gene expression analysis in NFI-C(-/-) and wild-type embryonic primary murine fibroblasts. This indicated that NFI-C acts mostly to repress gene expression in response to TGF-beta1. Misregulated genes were prominently overrepresented by regulators of connective tissue inflammation and repair. In vivo skin healing revealed a faster inflammatory stage and wound closure in NFI-C(-/-) mice. Expression of PDGFA and PDGF-receptor alpha were increased in wounds of NFI-C(-/-) mice, explaining the early recruitment of macrophages and fibroblasts. Differentiation of fibroblasts to contractile myofibroblasts was also elevated, providing a rationale for faster wound closure. Taken together with the role of TGF-beta in myofibroblast differentiation, our results imply a central role of NFI-C in the interplay of the two signaling pathways and in regulation of the progression of tissue regeneration.


Assuntos
Fatores de Transcrição NFI/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Pele/patologia , Fator de Crescimento Transformador beta1/metabolismo , Cicatrização , Animais , Diferenciação Celular , Movimento Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Macrófagos/citologia , Camundongos , Camundongos Knockout , Fatores de Transcrição NFI/deficiência , Análise de Sequência com Séries de Oligonucleotídeos , Reprodutibilidade dos Testes , Pele/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA