Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Cell Biol ; 22(6): 640-650, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32393888

RESUMO

Tissue homeostasis and regeneration rely on resident stem cells (SCs), whose behaviour is regulated through niche-dependent crosstalk. The mechanisms underlying SC identity are still unfolding. Here, using spatiotemporal gene ablation in murine hair follicles, we uncover a critical role for the transcription factors (TFs) nuclear factor IB (NFIB) and IX (NFIX) in maintaining SC identity. Without NFI TFs, SCs lose their hair-regenerating capability, and produce skin bearing striking resemblance to irreversible human alopecia, which also displays reduced NFIs. Through single-cell transcriptomics, ATAC-Seq and ChIP-Seq profiling, we expose a key role for NFIB and NFIX in governing super-enhancer maintenance of the key hair follicle SC-specific TF genes. When NFIB and NFIX are genetically removed, the stemness epigenetic landscape is lost. Super-enhancers driving SC identity are decommissioned, while unwanted lineages are de-repressed ectopically. Together, our findings expose NFIB and NFIX as crucial rheostats of tissue homeostasis, functioning to safeguard the SC epigenome from a breach in lineage confinement that otherwise triggers irreversible tissue degeneration.


Assuntos
Alopecia/patologia , Diferenciação Celular , Cromatina/metabolismo , Folículo Piloso/citologia , Fatores de Transcrição NFI/fisiologia , Células-Tronco/citologia , Alopecia/genética , Alopecia/metabolismo , Animais , Células Cultivadas , Cromatina/genética , Feminino , Folículo Piloso/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regeneração , Células-Tronco/metabolismo
2.
Neurosci Bull ; 36(7): 685-695, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32221845

RESUMO

Noxious mechanical information is transmitted through molecularly distinct nociceptors, with pinprick-evoked sharp sensitivity via A-fiber nociceptors marked by developmental expression of the neuropeptide Y receptor 2 (Npy2r) and von Frey filament-evoked punctate pressure information via unmyelinated C fiber nociceptors marked by MrgprD. However, the molecular programs controlling their development are only beginning to be understood. Here we demonstrate that Npy2r-expressing sensory neurons are in fact divided into two groups, based on transient or persistent Npy2r expression. Npy2r-transient neurons are myelinated, likely including A-fiber nociceptors, whereas Npy2r-persistent ones belong to unmyelinated pruriceptors that co-express Nppb. We then showed that the transcription factors NFIA and Runx1 are necessary for the development of Npy2r-transient A-fiber nociceptors and MrgprD+ C-fiber nociceptors, respectively. Behaviorally, mice with conditional knockout of Nfia, but not Runx1 showed a marked attenuation of pinprick-evoked nocifensive responses. Our studies therefore identify a transcription factor controlling the development of myelinated nociceptors.


Assuntos
Fatores de Transcrição NFI , Nociceptores , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Feminino , Gânglios Espinais/fisiologia , Masculino , Camundongos , Camundongos Knockout , Fatores de Transcrição NFI/fisiologia , Fibras Nervosas Amielínicas/fisiologia , Nociceptores/fisiologia , Receptores de Neuropeptídeo Y/fisiologia , Células Receptoras Sensoriais/fisiologia
3.
J Mol Histol ; 50(5): 471-482, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31432308

RESUMO

Nuclear factor I-C (NFIC) plays critical roles in the regulation of tooth development by influencing the biological behaviors of stem cells in the dental germ. This study aimed to investigate the effect of NFIC on the vitality and osteogenic/cementogenic differentiation of rat dental follicle cells (DFCs). DFCs were isolated from dental follicles in the first molars of neonatal rats. DFCs expressed mesenchymal stromal cell markers CD29, CD44 and CD90 and had capabilities for self-renewal and multipotent differentiation. Overexpression of NFIC promoted the proliferation of DFCs without markedly influencing the apoptosis of DFCs. Moreover, NFIC increased alkaline phosphatase (ALP) activity in DFCs and upregulated the mRNA levels of osteogenic-related markers, namely, collagen type I (Col I), Runt-related transcription factor 2 (Runx2) and ALP, as well as ß-catenin. In contrast, silencing NFIC by siRNA increased the apoptosis of DFCs and downregulated the expression of osteogenic-related markers. In conclusion, these results suggested that upregulation of NFIC may promote the proliferation and osteogenic/cementogenic differentiation of DFCs.


Assuntos
Saco Dentário/citologia , Fatores de Transcrição NFI/fisiologia , Osteogênese , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células-Tronco Mesenquimais , Dente Molar/citologia , Ratos
4.
Nat Commun ; 8(1): 1055, 2017 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-29057908

RESUMO

Muscular dystrophies are severe disorders due to mutations in structural genes, and are characterized by skeletal muscle wasting, compromised patient mobility, and respiratory functions. Although previous works suggested enhancing regeneration and muscle mass as therapeutic strategies, these led to no long-term benefits in humans. Mice lacking the transcription factor Nfix have delayed regeneration and a shift toward an oxidative fiber type. Here, we show that ablating or silencing the transcription factor Nfix ameliorates pathology in several forms of muscular dystrophy. Silencing Nfix in postnatal dystrophic mice, when the first signs of the disease already occurred, rescues the pathology and, conversely, Nfix overexpression in dystrophic muscles increases regeneration and markedly exacerbates the pathology. We therefore offer a proof of principle for a novel therapeutic approach for muscular dystrophies based on delaying muscle regeneration.


Assuntos
Músculos/fisiologia , Distrofias Musculares/genética , Fatores de Transcrição NFI/fisiologia , Regeneração , Animais , Feminino , Inativação Gênica , Masculino , Camundongos , Músculos/patologia , Distrofias Musculares/patologia , Sarcoglicanas/genética
5.
Cell Rep ; 14(9): 2238-2249, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26923583

RESUMO

Nfix belongs to a family of four highly conserved proteins that act as transcriptional activators and/or repressors of cellular and viral genes. We previously showed a pivotal role for Nfix in regulating the transcriptional switch from embryonic to fetal myogenesis. Here, we show that Nfix directly represses the Myostatin promoter, thus controlling the proper timing of satellite cell differentiation and muscle regeneration. Nfix-null mice display delayed regeneration after injury, and this deficit is reversed upon in vivo Myostatin silencing. Conditional deletion of Nfix in satellite cells results in a similar delay in regeneration, confirming the functional requirement for Nfix in satellite cells. Moreover, mice lacking Nfix show reduced myofiber cross sectional area and a predominant slow twitching phenotype. These data define a role for Nfix in postnatal skeletal muscle and unveil a mechanism for Myostatin regulation, thus providing insights into the modulation of its complex signaling pathway.


Assuntos
Fibras Musculares Esqueléticas/fisiologia , Miostatina/genética , Fatores de Transcrição NFI/fisiologia , Regeneração , Animais , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Inativação Gênica , Camundongos Transgênicos , Mioblastos/fisiologia , Miostatina/metabolismo
6.
J Leukoc Biol ; 99(2): 301-10, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26329426

RESUMO

Immune cell-lineage specification and function are influenced by progenitor origin and environmental factors. The mechanism of differentiation of immune cells, such as neutrophils, monocytes, and myeloid-derived suppressor cells, in inflammatory environments has not been elucidated completely. In this study, we have identified human microRNA-136 as a positive regulator of the differentiation of granulocytes and monocytes. Ectopic microRNA-136 induced cells to express higher levels of CD11b, CD14, and C/EBPε, secrete more cytokines, and synthesize higher levels of reactive oxygen species and H(2)O(2). microRNA-136 was shown to target and degrade multiple differentiation-associated molecules, such as the transcription factor NFIA, which induced the release of another microRNA, microRNA-223, with the ability to promote CD11b expression. Furthermore, microRNA-136 expression was remarkably increased by TNF-α, which activated NF-κB to bind to the DNA-promoter region controlling microRNA-136 expression. Additionally, TNF-α may alter NFIA expression through its modulation of microRNA-136 expression. Thus, TNF-α-mediated microRNA-136 may play a critical role in the generation and differentiation of inflammatory immune cells.


Assuntos
Regulação da Expressão Gênica/fisiologia , MicroRNAs/fisiologia , Células Mieloides/citologia , Mielopoese/fisiologia , Fatores de Transcrição NFI/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Regiões 3' não Traduzidas/genética , Antígeno CD11b/biossíntese , Antígeno CD11b/genética , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células HL-60 , Humanos , Peróxido de Hidrogênio/metabolismo , Receptores de Lipopolissacarídeos/biossíntese , Receptores de Lipopolissacarídeos/genética , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , MicroRNAs/genética , Células Mieloides/efeitos dos fármacos , Mielopoese/efeitos dos fármacos , NF-kappa B/metabolismo , Fatores de Transcrição NFI/biossíntese , Fatores de Transcrição NFI/genética , Oligonucleotídeos/genética , Regiões Promotoras Genéticas/genética , Espécies Reativas de Oxigênio/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
7.
J Orthop Res ; 33(8): 1205-11, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25764026

RESUMO

Giant cell tumor of bone (GCTb) represents 5% of bone tumors, and although considered benign, 5% metastasize to the lung. The expression of proteins directly or indirectly associated with osteolysis and tumor growth was studied on 163 samples of GCTb. Of these, 33 patients developed lung metastasis during follow-up. The impact of tumor-host interaction on clinical aspects was evaluated with the aim of finding specific markers for new biological therapies, thus improving clinical management of GCTb. Protein expression was evaluated by immunohistochemical analysis on Tissue Microarray. The majority of GCTb samples from patients with metastatic disease were strongly positive to RANKL and its receptor RANK as well as to CAII and MMP-2 and to pro-survival proteins NFIB and c-Fos. Kaplan-Meier analysis indicated a significant difference in metastasis free survival curves based on protein staining. Interestingly, the statistical correlation established a strong association between all variables studied with a higher τ coefficient for RANK/RANKL, RANK/NFIB, and RANKL/NFIB pairs. At multivariate analysis co-overexpression of NFIB, RANK and RANKL significantly increased the risk of metastasis with an odds ratio of 13.59 (95%CI 4.12-44.82; p < 0.0005). In conclusion, the interconnection between matrix remodeling and tumor cell activity may identify tumor-host endpoints for new biological treatments.


Assuntos
Neoplasias Ósseas/mortalidade , Tumor de Células Gigantes do Osso/mortalidade , Fatores de Transcrição NFI/fisiologia , Adulto , Idoso , Neoplasias Ósseas/química , Neoplasias Ósseas/patologia , Remodelação Óssea , Feminino , Tumor de Células Gigantes do Osso/química , Tumor de Células Gigantes do Osso/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Ligante RANK/análise , Receptor Ativador de Fator Nuclear kappa-B/análise , Estudos Retrospectivos
8.
Exp Cell Res ; 332(2): 259-66, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25668322

RESUMO

The transcription factor Nuclear Factor I-C (NFIC) has been implicated in the regulation of tooth root development, where it may be anticipated to impact on the behavior of stem cells from the apical papilla (SCAPs) and root odontoblast activity. We hypothesized that NFIC may provide an important target for promoting dentin/root regeneration. In the present study, the effects of NFIC on the proliferation and differentiation of SCAPs were investigated. Over-expression of NFIC increased cell proliferation, mineralization nodule formation and alkaline phosphatase (ALP) activity in SCAPs. Furthermore, NFIC up-regulated the mRNA levels of odontogenic-related markers, ALP, osteocalcin and collagen type I as well as dentin sialoprotein protein levels. In contrast, knockdown of NFIC by si-RNA inhibited the mineralization capacity of SCAPs and down-regulated the expression of odontogenic-related markers. In conclusion, the results indicated that upregulation of NFIC activity in SCAPs may promote osteo/odontoblastic differentiation of SCAPs.


Assuntos
Células-Tronco Adultas/fisiologia , Diferenciação Celular , Proliferação de Células , Papila Dentária/citologia , Fatores de Transcrição NFI/fisiologia , Adolescente , Fosfatase Alcalina/metabolismo , Células Cultivadas , Humanos , Calcificação de Dente
9.
J Dent Res ; 94(2): 312-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25403566

RESUMO

NFIB (nuclear factor I B) is a NFI transcription factor family member, which is essential for the development of a variety of organ systems. Salivary gland development occurs through several stages, including prebud, bud, pseudoglandular, canalicular, and terminal. Although many studies have been done to understand mouse submandibular gland (SMG) branching morphogenesis, little is known about SMG cell differentiation during the terminal stages. The goal of this study was to determine the role of NFIB during SMG development. We analyzed SMGs from wild-type and Nfib-deficient mice (Nfib (-/-)). At embryonic (E) day 18.5, SMGs from wild-type mice showed duct branching morphogenesis and differentiation of tubule ductal cells into tubule secretory cells. In contrast, SMGs from Nfib (-/-) mice at E18.5 failed to differentiate into tubule secretory cells while branching morphogenesis was unaffected. SMGs from wild-type mice at E16.5 displayed well-organized cuboidal inner terminal tubule cells. However, SMGs from Nfib (-/-) at E16.5 displayed disorganized inner terminal tubule cells. SMGs from wild-type mice at E18.5 became fully differentiated, as indicated by a high degree of apicobasal polarization (i.e., presence of apical ZO-1 and basolateral E-cadherin) and columnar shape. Furthermore, SMGs from wild-type mice at E18.5 expressed the protein SMGC, a marker for tubule secretory cells. However, SMGs from Nfib (-/-) mice at E18.5 showed apicobasal polarity, but they were disorganized and lost the ability to secrete SMGC. These findings indicate that the transcription factor NFIB is not required for branching morphogenesis but plays a key role in tubule cell differentiation during mouse SMG development.


Assuntos
Fatores de Transcrição NFI/fisiologia , Glândula Submandibular/embriologia , Animais , Aquaporina 5/análise , Biomarcadores/análise , Caderinas/análise , Diferenciação Celular/fisiologia , Polaridade Celular/fisiologia , Forma Celular , Desenvolvimento Embrionário , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Morfogênese/fisiologia , Mucinas/análise , Fatores de Transcrição NFI/genética , Ductos Salivares/citologia , Ductos Salivares/embriologia , Glândula Submandibular/citologia , Proteína da Zônula de Oclusão-1/análise
10.
Cereb Cortex ; 25(10): 3758-78, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25331604

RESUMO

Transcription factors of the nuclear factor one (NFI) family play a pivotal role in the development of the nervous system. One member, NFIX, regulates the development of the neocortex, hippocampus, and cerebellum. Postnatal Nfix(-/-) mice also display abnormalities within the subventricular zone (SVZ) lining the lateral ventricles, a region of the brain comprising a neurogenic niche that provides ongoing neurogenesis throughout life. Specifically, Nfix(-/-) mice exhibit more PAX6-expressing progenitor cells within the SVZ. However, the mechanism underlying the development of this phenotype remains undefined. Here, we reveal that NFIX contributes to multiple facets of SVZ development. Postnatal Nfix(-/-) mice exhibit increased levels of proliferation within the SVZ, both in vivo and in vitro as assessed by a neurosphere assay. Furthermore, we show that the migration of SVZ-derived neuroblasts to the olfactory bulb is impaired, and that the olfactory bulbs of postnatal Nfix(-/-) mice are smaller. We also demonstrate that gliogenesis within the rostral migratory stream is delayed in the absence of Nfix, and reveal that Gdnf (glial-derived neurotrophic factor), a known attractant for SVZ-derived neuroblasts, is a target for transcriptional activation by NFIX. Collectively, these findings suggest that NFIX regulates both proliferation and migration during the development of the SVZ neurogenic niche.


Assuntos
Movimento Celular , Proliferação de Células , Ventrículos Laterais/embriologia , Fatores de Transcrição NFI/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese , Animais , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Interneurônios/fisiologia , Ventrículos Laterais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Neuroglia/fisiologia , Bulbo Olfatório/embriologia , Bulbo Olfatório/metabolismo , Nicho de Células-Tronco
11.
Mol Endocrinol ; 28(5): 758-67, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24678731

RESUMO

Mammary-specific genetic programs are activated during pregnancy by the common transcription factor signal transducer and activator of transcription (STAT) 5. More than one third of these genes carry nuclear factor I/B (NFIB) binding motifs that coincide with STAT5 in vivo binding, suggesting functional synergy between these two transcription factors. The role of NFIB in this governance was investigated in mice from which Nfib had been inactivated in mammary stem cells or in differentiating alveolar epithelium. Although NFIB was not required for alveolar expansion, the combined absence of NFIB and STAT5 prevented the formation of functional alveoli. NFIB controlled the expression of mammary-specific and STAT5-regulated genes and chromatin immunoprecipitation-sequencing established STAT5 and NFIB binding at composite regulatory elements containing histone H3 lysine dimethylation enhancer marks and progesterone receptor binding. By integrating previously published chromatin immunoprecipitation-sequencing data sets, the presence of NFIB-STAT5 modules in other cell types was investigated. Notably, genomic sites bound by NFIB in hair follicle stem cells were also occupied by STAT5 in mammary epithelium and coincided with enhancer marks. Many of these genes were under NFIB control in both hair follicle stem cells and mammary alveolar epithelium. We propose that NFIB-STAT5 modules, possibly in conjunction with other transcription factors, control cell-specific genetic programs.


Assuntos
Fatores de Transcrição NFI/fisiologia , Fator de Transcrição STAT5/fisiologia , Animais , Sequência de Bases , Sítios de Ligação , Sequência Consenso , Feminino , Regulação da Expressão Gênica , Ontologia Genética , Lactação , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/fisiologia , Camundongos Nus , Camundongos Transgênicos , Gravidez , Transcriptoma
12.
J Neurosci ; 34(8): 2921-30, 2014 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-24553933

RESUMO

Epigenetic mechanisms are essential in regulating neural progenitor cell self-renewal, with the chromatin-modifying protein Enhancer of zeste homolog 2 (EZH2) emerging as a central player in promoting progenitor cell self-renewal during cortical development. Despite this, how Ezh2 is itself regulated remains unclear. Here, we demonstrate that the transcription factor nuclear factor IB (NFIB) plays a key role in this process. Nfib(-/-) mice exhibit an increased number of proliferative ventricular zone cells that express progenitor cell markers and upregulation of EZH2 expression within the neocortex and hippocampus. NFIB binds to the Ezh2 promoter and overexpression of NFIB represses Ezh2 transcription. Finally, key downstream targets of EZH2-mediated epigenetic repression are misregulated in Nfib(-/-) mice. Collectively, these results suggest that the downregulation of Ezh2 transcription by NFIB is an important component of the process of neural progenitor cell differentiation during cortical development.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Epigênese Genética/fisiologia , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/fisiologia , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/fisiologia , Animais , Contagem de Células , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Ensaio de Desvio de Mobilidade Eletroforética , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Análise em Microsséries , Mutação/genética , Mutação/fisiologia , Células-Tronco Neurais/fisiologia , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase em Tempo Real
13.
Cereb Cortex ; 24(1): 261-79, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23042739

RESUMO

Neural progenitor cells have the ability to give rise to neurons and glia in the embryonic, postnatal and adult brain. During development, the program regulating whether these cells divide and self-renew or exit the cell cycle and differentiate is tightly controlled, and imbalances to the normal trajectory of this process can lead to severe functional consequences. However, our understanding of the molecular regulation of these fundamental events remains limited. Moreover, processes underpinning development of the postnatal neurogenic niches within the cortex remain poorly defined. Here, we demonstrate that Nuclear factor one X (NFIX) is expressed by neural progenitor cells within the embryonic hippocampus, and that progenitor cell differentiation is delayed within Nfix(-/-) mice. Moreover, we reveal that the morphology of the dentate gyrus in postnatal Nfix(-/-) mice is abnormal, with fewer subgranular zone neural progenitor cells being generated in the absence of this transcription factor. Mechanistically, we demonstrate that the progenitor cell maintenance factor Sry-related HMG box 9 (SOX9) is upregulated in the hippocampus of Nfix(-/-) mice and demonstrate that NFIX can repress Sox9 promoter-driven transcription. Collectively, our findings demonstrate that NFIX plays a central role in hippocampal morphogenesis, regulating the formation of neuronal and glial populations within this structure.


Assuntos
Diferenciação Celular/fisiologia , Hipocampo/embriologia , Fatores de Transcrição NFI/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Contagem de Células , Corantes , Biologia Computacional , Giro Denteado/embriologia , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/fisiologia , Ensaio de Desvio de Mobilidade Eletroforética , Eletroporação , Feminino , Hematoxilina , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Luciferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/metabolismo , Inclusão em Parafina , Gravidez , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase em Tempo Real
15.
Development ; 140(7): 1528-36, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23482488

RESUMO

During mouse skeletal muscle development, the Nfix gene has a pivotal role in regulating fetal-specific transcription. Zebrafish and mice share related programs for muscle development, although zebrafish develops at a much faster rate. In fact, although mouse fetal muscle fibers form after 15 days of development, in fish secondary muscle fibers form by 48 hours post-fertilization in a process that until now has been poorly characterized mechanically. In this work, we studied the zebrafish ortholog Nfix (nfixa) and its role in the proper switch to the secondary myogenic wave. This allowed us to highlight evolutionarily conserved and divergent functions of Nfix. In fact, the knock down of nfixa in zebrafish blocks secondary myogenesis, as in mouse, but also alters primary slow muscle fiber formation. Moreover, whereas Nfix mutant mice are motile, nfixa knockdown zebrafish display impaired motility that probably depends upon disruption of the sarcoplasmic reticulum. We conclude that, during vertebrate evolution, the transcription factor Nfix lost some specific functions, probably as a consequence of the different environment in which teleosts and mammals develop.


Assuntos
Evolução Molecular , Músculo Esquelético/embriologia , Fatores de Transcrição NFI/fisiologia , Vertebrados/genética , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Sequência Conservada/fisiologia , Embrião de Mamíferos , Embrião não Mamífero , Especiação Genética , Camundongos , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Fatores de Transcrição NFI/genética , Filogenia , Vertebrados/embriologia , Vertebrados/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
16.
J Neurosci ; 33(7): 2860-72, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23407945

RESUMO

Dendrite and synapse development are critical for establishing appropriate neuronal circuits, and disrupted timing of these events can alter neural connectivity. Using microarrays, we have identified a nuclear factor I (NFI)-regulated temporal switch program linked to dendrite formation in developing mouse cerebellar granule neurons (CGNs). NFI function was required for upregulation of many synapse-related genes as well as downregulation of genes expressed in immature CGNs. Chromatin immunoprecipitation analysis revealed that a central feature of this program was temporally regulated NFI occupancy of late-expressed gene promoters. Developing CGNs undergo a hyperpolarizing shift in membrane potential, and depolarization inhibits their dendritic and synaptic maturation via activation of calcineurin (CaN) (Okazawa et al., 2009). Maintaining immature CGNs in a depolarized state blocked NFI temporal occupancy of late-expressed genes and the NFI switch program via activation of the CaN/nuclear factor of activated T-cells, cytoplasmic (NFATc) pathway and promotion of late-gene occupancy by NFATc4, and these mechanisms inhibited dendritogenesis. Conversely, inhibition of the CaN/NFATc pathway in CGNs maturing under physiological nondepolarizing conditions upregulated the NFI switch program, NFI temporal occupancy, and dendrite formation. NFATc4 occupied the promoters of late-expressed NFI program genes in immature mouse cerebellum, and its binding was temporally downregulated with development. Further, NFI temporal binding and switch gene expression were upregulated in the developing cerebellum of Nfatc4 (-/-) mice. These findings define a novel NFI switch and temporal occupancy program that forms a critical link between membrane potential/CaN and dendritic maturation in CGNs. CaN inhibits the program and NFI occupancy in immature CGNs by promoting NFATc4 binding to late-expressed genes. As maturing CGNs become more hyperpolarized, NFATc4 binding declines leading to onset of NFI temporal binding and the NFI switch program.


Assuntos
Calcineurina/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFI/fisiologia , Neurônios/fisiologia , Animais , Canais de Cálcio Tipo L/metabolismo , Diferenciação Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Biologia Computacional , Citoplasma/metabolismo , Dendritos/fisiologia , Feminino , Imunofluorescência , Vetores Genéticos , Lentivirus/genética , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Análise em Microsséries , Fatores de Transcrição NFI/biossíntese , Fatores de Transcrição NFI/genética , Plasmídeos/genética , Linfócitos T/metabolismo , Imagens com Corantes Sensíveis à Voltagem
17.
Cerebellum ; 11(1): 41-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22548229

RESUMO

Granule neurons have a central role in cerebellar function via their synaptic interactions with other neuronal cell types both within and outside this structure. Establishment of these synaptic connections and its control is therefore essential to their function. Both intrinsic as well as environmental mechanisms are required for neuronal development and formation of neuronal circuits, and a key but poorly understood question is how these various events are coordinated and integrated in maturing neurons. In this review, we summarize recent work on the role of the Nuclear Factor I family in the transcriptional programming of cerebellar granule neuron maturation and synapse formation. In particular, we describe (1) the involvement of this family of factors in key developmental steps occurring throughout postmitotic granule neuron development, including dendrite and synapse formation and synaptic receptor expression, and (2) the mediation of these actions by critical downstream gene targets that control cell-cell interactions. These findings illustrate how Nuclear Factor I proteins and their regulons function as a "bridge" between cell-intrinsic and cell-extrinsic interactions to control multiple phases of granule neuron development.


Assuntos
Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Córtex Cerebelar/crescimento & desenvolvimento , Córtex Cerebelar/metabolismo , Grânulos Citoplasmáticos/metabolismo , Fatores de Transcrição NFI/fisiologia , Neurogênese/fisiologia , Neurônios/metabolismo , Animais , Córtex Cerebelar/citologia , Grânulos Citoplasmáticos/fisiologia , Humanos , Fatores de Transcrição NFI/metabolismo , Vias Neurais/citologia , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Neurônios/citologia , Neurônios/fisiologia
18.
Neuron ; 74(1): 79-94, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22500632

RESUMO

Transcriptional cascades that operate over the course of lineage development are fundamental mechanisms that control cellular differentiation. In the developing central nervous system (CNS), these mechanisms are well characterized during neurogenesis, but remain poorly defined during neural stem cell commitment to the glial lineage. NFIA is a transcription factor that plays a crucial role in the onset of gliogenesis; we found that its induction is regulated by the transcription factor Sox9 and that this relationship mediates the initiation of gliogenesis. Subsequently, Sox9 and NFIA form a complex and coregulate a set of genes induced after glial initiation. Functional studies revealed that a subset of these genes, Apcdd1 and Mmd2, perform key migratory and metabolic roles during astro-gliogenesis, respectively. In sum, these studies delineate a transcriptional regulatory cascade that operates during the initiation of gliogenesis and identifies a unique set of genes that regulate key aspects of astro-glial precursor physiology during development.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Transcrição NFI/fisiologia , Neuroglia/citologia , Fatores de Transcrição SOX9/fisiologia , Animais , Linhagem da Célula/fisiologia , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Embrião de Galinha , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Camundongos , Neuroglia/fisiologia , Organogênese/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Transcrição Gênica
19.
J Comp Neurol ; 519(17): 3532-48, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21800304

RESUMO

Development of the cerebellum involves the coordinated proliferation, differentiation, maturation, and integration of cells from multiple neuronal and glial lineages. In rodent models, much of this occurs in the early postnatal period. However, our understanding of the molecular mechanisms that regulate this phase of cerebellar development remains incomplete. Here, we address the role of the transcription factor nuclear factor one X (NFIX), in postnatal development of the cerebellum. NFIX is expressed by progenitor cells within the external granular layer and by cerebellar granule neurons within the internal granule layer. Using NFIX⁻/⁻ mice, we demonstrate that the development of cerebellar granule neurons and Purkinje cells within the postnatal cerebellum is delayed in the absence of this transcription factor. Furthermore, the differentiation of mature glia within the cerebellum, such as Bergmann glia, is also significantly delayed in the absence of NFIX. Collectively, the expression pattern of NFIX, coupled with the delays in the differentiation of multiple cell populations of the developing cerebellum in NFIX⁻/⁻ mice, suggest a central role for NFIX in the regulation of cerebellar development, highlighting the importance of this gene for the maturation of this key structure.


Assuntos
Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Fatores de Transcrição NFI/fisiologia , Neurogênese/fisiologia , Células-Tronco/fisiologia , Animais , Animais Recém-Nascidos , Grânulos Citoplasmáticos/química , Grânulos Citoplasmáticos/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
20.
Proc Natl Acad Sci U S A ; 108(27): E265-74, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690374

RESUMO

The sequential production of neurons and astrocytes from neuroepithelial precursors is a fundamental feature of central nervous system development. We report that LIM-homeodomain (LIM-HD) transcription factor Lhx2 regulates this transition in the developing hippocampus. Disrupting Lhx2 function in the embryonic hippocampus by in utero electroporation and in organotypic slice culture caused the premature production of astrocytes at stages when neurons are normally generated. Lhx2 function is therefore necessary to suppress astrogliogenesis during the neurogenic period. Furthermore, Lhx2 overexpression was sufficient to suppress astrogliogenesis and prolong the neurogenic period. We provide evidence that Lhx2 overexpression can counteract the instructive astrogliogenic effect of Notch activation. Lhx2 overexpression was also able to override and suppress the activation of the GFAP promoter by Nfia, a Notch-regulated transcription factor that is required for gliogenesis. Thus, Lhx2 appears to act as a "brake" on Notch/Nfia-mediated astrogliogenesis. This critical role for Lhx2 is spatially restricted to the hippocampus, because loss of Lhx2 function in the neocortex did not result in premature astrogliogenesis at the expense of neurogenesis. Our results therefore place Lhx2 as a central regulator of the neuron-glia cell fate decision in the hippocampus and reveal a striking regional specificity of this fundamental function within the dorsal telencephalon.


Assuntos
Hipocampo/embriologia , Proteínas de Homeodomínio/fisiologia , Neurogênese/fisiologia , Fatores de Transcrição/fisiologia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição NFI/fisiologia , Neocórtex/citologia , Neocórtex/embriologia , Neocórtex/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Fenótipo , Gravidez , Receptores Notch/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...