Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Cell ; 140(4): 554-66, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20178747

RESUMO

Skeletal myogenesis, like hematopoiesis, occurs in successive developmental stages that involve different cell populations and expression of different genes. We show here that the transcription factor nuclear factor one X (Nfix), whose expression is activated by Pax7 in fetal muscle, in turn activates the transcription of fetal specific genes such as MCK and beta-enolase while repressing embryonic genes such as slow myosin. In the case of the MCK promoter, Nfix forms a complex with PKC theta that binds, phosphorylates, and activates MEF2A. Premature expression of Nfix activates fetal and suppresses embryonic genes in embryonic muscle, whereas muscle-specific ablation of Nfix prevents fetal and maintains embryonic gene expression in the fetus. Therefore, Nfix acts as a transcriptional switch from embryonic to fetal myogenesis.


Assuntos
Desenvolvimento Muscular , Músculo Esquelético/embriologia , Fatores de Transcrição NFI/metabolismo , Transcrição Gênica , Animais , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Isoenzimas/metabolismo , Fatores de Transcrição MEF2 , Camundongos , Fatores de Regulação Miogênica/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fator de Transcrição PAX7/metabolismo , Fosfopiruvato Hidratase , Proteína Quinase C/metabolismo , Proteína Quinase C-theta
2.
Biol Reprod ; 106(6): 1191-1205, 2022 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-35243487

RESUMO

Members of the nuclear factor I (NFI) family are key regulators of stem cell biology during development, with well-documented roles for NFIA, NFIB, and NFIX in a variety of developing tissues, including brain, muscle, and lung. Given the central role these factors play in stem cell biology, we posited that they may be pivotal for spermatogonial stem cells or further developing spermatogonia during testicular development. Surprisingly, in stark contrast to other developing organ systems where NFI members are co-expressed, these NFI family members show discrete patterns of expression within the seminiferous tubules. Sertoli cells (spermatogenic supporting cells) express NFIA, spermatocytes express NFIX, round spermatids express NFIB, and peritubular myoid cells express each of these three family members. Further analysis of NFIX expression during the cycle of the seminiferous epithelium revealed expression not in spermatogonia, as we anticipated, but in spermatocytes. These data suggested a potential role for NFIX in spermatogenesis. To investigate, we analyzed mice with constitutive deletion of Nfix (Nfix-null). Assessment of germ cells in the postnatal day 20 (P20) testes of Nfix-null mice revealed that spermatocytes initiate meiosis, but zygotene stage spermatocytes display structural defects in the synaptonemal complex, and increased instances of unrepaired DNA double-strand breaks. Many developing spermatocytes in the Nfix-null testis exhibited multinucleation. As a result of these defects, spermatogenesis is blocked at early diplotene and very few round spermatids are produced. Collectively, these novel data establish the global requirement for NFIX in correct meiotic progression during the first wave of spermatogenesis.


Assuntos
Fatores de Transcrição NFI , Espermatogônias , Testículo , Animais , Masculino , Meiose , Camundongos , Camundongos Knockout , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Espermatócitos/metabolismo , Espermatogênese/genética , Testículo/metabolismo
3.
Carcinogenesis ; 42(3): 357-368, 2021 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-33346791

RESUMO

Nuclear factor one (NFI) transcription factors are implicated in both brain development and cancer in mice and humans and play an essential role in glial differentiation. NFI expression is reduced in human astrocytoma samples, particularly those of higher grade, whereas over-expression of NFI protein can induce the differentiation of glioblastoma cells within human tumour xenografts and in glioblastoma cell lines in vitro. These data indicate that NFI proteins may act as tumour suppressors in glioma. To test this hypothesis, we generated complex mouse genetic crosses involving six alleles to target gene deletion of known tumour suppressor genes that induce endogenous high-grade glioma in mice, and overlaid this with loss of function Nfi mutant alleles, Nfia and Nfib, a reporter transgene and an inducible Cre allele. Deletion of Nfi resulted in reduced survival time of the mice, increased tumour load and a more aggressive tumour phenotype than observed in glioma mice with normal expression of NFI. Together, these data indicate that NFI genes represent a credible target for both diagnostic analyses and therapeutic strategies to combat high-grade glioma.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Fatores de Transcrição NFI/metabolismo , Animais , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Fatores de Transcrição NFI/genética
4.
Am J Hum Genet ; 103(5): 752-768, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30388402

RESUMO

The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.


Assuntos
Haploinsuficiência/genética , Deficiência Intelectual/genética , Megalencefalia/genética , Fatores de Transcrição NFI/genética , Adolescente , Adulto , Animais , Córtex Cerebral/patologia , Criança , Pré-Escolar , Códon sem Sentido/genética , Estudos de Coortes , Corpo Caloso/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Polimorfismo de Nucleotídeo Único/genética , Adulto Jovem
5.
Development ; 145(3)2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29437824

RESUMO

Our understanding of the transcriptional programme underpinning adult hippocampal neurogenesis is incomplete. In mice, under basal conditions, adult hippocampal neural stem cells (AH-NSCs) generate neurons and astrocytes, but not oligodendrocytes. The factors limiting oligodendrocyte production, however, remain unclear. Here, we reveal that the transcription factor NFIX plays a key role in this process. NFIX is expressed by AH-NSCs, and its expression is sharply upregulated in adult hippocampal neuroblasts. Conditional ablation of Nfix from AH-NSCs, coupled with lineage tracing, transcriptomic sequencing and behavioural studies collectively reveal that NFIX is cell-autonomously required for neuroblast maturation and survival. Moreover, a small number of AH-NSCs also develop into oligodendrocytes following Nfix deletion. Remarkably, when Nfix is deleted specifically from intermediate progenitor cells and neuroblasts using a Dcx-creERT2 driver, these cells also display elevated signatures of oligodendrocyte gene expression. Together, these results demonstrate the central role played by NFIX in neuroblasts within the adult hippocampal stem cell neurogenic niche in promoting the maturation and survival of these cells, while concomitantly repressing oligodendrocyte gene expression signatures.


Assuntos
Hipocampo/citologia , Hipocampo/metabolismo , Fatores de Transcrição NFI/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular , Proteína Duplacortina , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/crescimento & desenvolvimento , Masculino , Transtornos da Memória/genética , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Knockout , Fatores de Transcrição NFI/deficiência , Fatores de Transcrição NFI/genética , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/fisiologia , Regulação para Cima
6.
Genes Dev ; 27(16): 1769-86, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23964093

RESUMO

The majority of neural stem cells (NSCs) in the adult brain are quiescent, and this fraction increases with aging. Although signaling pathways that promote NSC quiescence have been identified, the transcriptional mechanisms involved are mostly unknown, largely due to lack of a cell culture model. In this study, we first demonstrate that NSC cultures (NS cells) exposed to BMP4 acquire cellular and transcriptional characteristics of quiescent cells. We then use epigenomic profiling to identify enhancers associated with the quiescent NS cell state. Motif enrichment analysis of these enhancers predicts a major role for the nuclear factor one (NFI) family in the gene regulatory network controlling NS cell quiescence. Interestingly, we found that the family member NFIX is robustly induced when NS cells enter quiescence. Using genome-wide location analysis and overexpression and silencing experiments, we demonstrate that NFIX has a major role in the induction of quiescence in cultured NSCs. Transcript profiling of NS cells overexpressing or silenced for Nfix and the phenotypic analysis of the hippocampus of Nfix mutant mice suggest that NFIX controls the quiescent state by regulating the interactions of NSCs with their microenvironment.


Assuntos
Epigênese Genética , Fatores de Transcrição NFI/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Animais , Proteína Morfogenética Óssea 4/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Elementos Facilitadores Genéticos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/efeitos dos fármacos , Ligação Proteica
7.
Cerebellum ; 19(1): 89-101, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31838646

RESUMO

Transcriptional regulation plays a central role in controlling neural stem and progenitor cell proliferation and differentiation during neurogenesis. For instance, transcription factors from the nuclear factor I (NFI) family have been shown to co-ordinate neural stem and progenitor cell differentiation within multiple regions of the embryonic nervous system, including the neocortex, hippocampus, spinal cord and cerebellum. Knockout of individual Nfi genes culminates in similar phenotypes, suggestive of common target genes for these transcription factors. However, whether or not the NFI family regulates common suites of genes remains poorly defined. Here, we use granule neuron precursors (GNPs) of the postnatal murine cerebellum as a model system to analyse regulatory targets of three members of the NFI family: NFIA, NFIB and NFIX. By integrating transcriptomic profiling (RNA-seq) of Nfia- and Nfix-deficient GNPs with epigenomic profiling (ChIP-seq against NFIA, NFIB and NFIX, and DNase I hypersensitivity assays), we reveal that these transcription factors share a large set of potential transcriptional targets, suggestive of complementary roles for these NFI family members in promoting neural development.


Assuntos
Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Fatores de Transcrição NFI/metabolismo , Animais , Animais Recém-Nascidos , Cerebelo/citologia , Sequenciamento de Cromatina por Imunoprecipitação/métodos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição NFI/genética , Neurogênese/fisiologia , Gravidez
8.
Cereb Cortex ; 29(8): 3590-3604, 2019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-30272140

RESUMO

Understanding the migration of newborn neurons within the brain presents a major challenge in contemporary biology. Neuronal migration is widespread within the developing brain but is also important within the adult brain. For instance, stem cells within the ventricular-subventricular zone (V-SVZ) and the subgranular zone of dentate gyrus of the adult rodent brain produce neuroblasts that migrate to the olfactory bulb and granule cell layer of the dentate gyrus, respectively, where they regulate key brain functions including innate olfactory responses, learning, and memory. Critically, our understanding of the factors mediating neuroblast migration remains limited. The transcription factor nuclear factor I X (NFIX) has previously been implicated in embryonic cortical development. Here, we employed conditional ablation of Nfix from the adult mouse brain and demonstrated that the removal of this gene from either neural stem and progenitor cells, or neuroblasts, within the V-SVZ culminated in neuroblast migration defects. Mechanistically, we identified aberrant neuroblast branching, due in part to increased expression of the guanylyl cyclase natriuretic peptide receptor 2 (Npr2), as a factor contributing to abnormal migration in Nfix-deficient adult mice. Collectively, these data provide new insights into how neuroblast migration is regulated at a transcriptional level within the adult brain.


Assuntos
Movimento Celular/genética , Giro Denteado/citologia , Ventrículos Laterais/citologia , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Camundongos , Células-Tronco Neurais/citologia , Neurogênese/genética , Receptores do Fator Natriurético Atrial/genética
9.
Am J Med Genet C Semin Med Genet ; 181(4): 611-626, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31730271

RESUMO

The nuclear factor one (NFI) site-specific DNA-binding proteins represent a family of transcription factors that are important for the development of multiple organ systems, including the brain. During brain development in mice, the expression patterns of Nfia, Nfib, and Nfix overlap, and knockout mice for each of these exhibit overlapping brain defects, including megalencephaly, dysgenesis of the corpus callosum, and enlarged ventricles, which implies a common but not redundant function in brain development. In line with these models, human phenotypes caused by haploinsufficiency of NFIA, NFIB, and NFIX display significant overlap, sharing neurodevelopmental deficits, macrocephaly, brain anomalies, and variable somatic overgrowth. Other anomalies may be present depending on the NFI gene involved. The possibility of variants in NFI genes should therefore be considered in individuals with intellectual disability and brain overgrowth, with individual NFI-related conditions being differentiated from one another by additional signs and symptoms. The exception is provided by specific NFIX variants that act in a dominant negative manner, as these cause a recognizable entity with more severe cognitive impairment and marked bone dysplasia, Marshall-Smith syndrome. NFIX duplications are associated with a phenotype opposite to that of haploinsufficiency, characterized by short stature, small head circumference, and delayed bone age. The spectrum of NFI-related disorders will likely be further expanded, as larger cohorts are assessed.


Assuntos
Crescimento/genética , Mutação , Fatores de Transcrição NFI/genética , Anormalidades Múltiplas/genética , Animais , Doenças do Desenvolvimento Ósseo/genética , Anormalidades Craniofaciais/genética , Duplicação Gênica , Transtornos do Crescimento/genética , Humanos , Camundongos , Displasia Septo-Óptica/genética , Síndrome
10.
Development ; 143(24): 4620-4630, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27965439

RESUMO

During forebrain development, radial glia generate neurons through the production of intermediate progenitor cells (IPCs). The production of IPCs is a central tenet underlying the generation of the appropriate number of cortical neurons, but the transcriptional logic underpinning this process remains poorly defined. Here, we examined IPC production using mice lacking the transcription factor nuclear factor I/X (Nfix). We show that Nfix deficiency delays IPC production and prolongs the neurogenic window, resulting in an increased number of neurons in the postnatal forebrain. Loss of additional Nfi alleles (Nfib) resulted in a severe delay in IPC generation while, conversely, overexpression of NFIX led to precocious IPC generation. Mechanistically, analyses of microarray and ChIP-seq datasets, coupled with the investigation of spindle orientation during radial glial cell division, revealed that NFIX promotes the generation of IPCs via the transcriptional upregulation of inscuteable (Insc). These data thereby provide novel insights into the mechanisms controlling the timely transition of radial glia into IPCs during forebrain development.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Hipocampo/embriologia , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/citologia , Neurogênese/genética , Animais , Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Neurogênese/fisiologia , Neurônios/citologia , Regiões Promotoras Genéticas/genética , Transcrição Gênica , Ativação Transcricional/genética
11.
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
12.
Dev Biol ; 432(2): 286-297, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29106906

RESUMO

During mouse spinal cord development, ventricular zone progenitor cells transition from producing neurons to producing glia at approximately embryonic day 11.5, a process known as the gliogenic switch. The transcription factors Nuclear Factor I (NFI) A and B initiate this developmental transition, but the contribution of a third NFI member, NFIX, remains unknown. Here, we reveal that ventricular zone progenitor cells within the spinal cord express NFIX after the onset of NFIA and NFIB expression, and after the gliogenic switch has occurred. Mice lacking NFIX exhibit normal neurogenesis within the spinal cord, and, while early astrocytic differentiation proceeds normally, aspects of terminal astrocytic differentiation are impaired. Finally, we report that, in the absence of Nfia or Nfib, there is a marked reduction in the spinal cord expression of NFIX, and that NFIB can transcriptionally activate Nfix expression in vitro. These data demonstrate that NFIX is part of the downstream transcriptional program through which NFIA and NFIB coordinate gliogenesis within the spinal cord. This hierarchical organisation of NFI protein expression and function during spinal cord gliogenesis reveals a previously unrecognised auto-regulatory mechanism within this gene family.


Assuntos
Fatores de Transcrição NFI/metabolismo , Medula Espinal/embriologia , Animais , Astrócitos/metabolismo , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFI/genética , Neurogênese , Neuroglia/metabolismo , Neurônios/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo , Células-Tronco/metabolismo , Ativação Transcricional
14.
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
15.
Dev Dyn ; 244(3): 227-38, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25156673

RESUMO

Nuclear factor one (NFI) transcription factors are a group of site-specific DNA-binding proteins that are emerging as critical regulators of stem cell biology. During development NFIs promote the production of differentiated progeny at the expense of stem cell fate, with Nfi null mice exhibiting defects such as severely delayed brain and lung maturation, skeletomuscular defects and renal abnormalities, phenotypes that are often consistent with patients with congenital Nfi mutations. Intriguingly, recent research suggests that in adult tissues NFI factors play a qualitatively different role than during development, with NFIs serving to promote the survival and maintenance of slow-cycling adult stem cell populations rather than their differentiation. Here we review the role of NFI factors in development, largely focusing on their role as promoters of stem cell differentiation, and attempt to reconcile this with the emerging role of NFIs in adult stem cell niches.


Assuntos
Células-Tronco Adultas/metabolismo , Diferenciação Celular/fisiologia , Fatores de Transcrição NFI/metabolismo , Células-Tronco Adultas/citologia , Animais , Sobrevivência Celular/fisiologia , Humanos , Camundongos , Camundongos Mutantes , Fatores de Transcrição NFI/genética
16.
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
17.
Stem Cells ; 32(9): 2467-79, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24801901

RESUMO

In bone marrow, bone marrow stromal cells (BMSCs) have the capacity to differentiate into osteoblasts and adipocytes. Age-related osteoporosis is associated with a reciprocal decrease of osteogenesis and an increase of adipogenesis in bone marrow. In this study, we demonstrate that disruption of nuclear factor I-C (NFI-C) impairs osteoblast differentiation and bone formation, and increases bone marrow adipocytes. Interestingly, NFI-C controls postnatal bone formation but does not influence prenatal bone development. We also found decreased NFI-C expression in osteogenic cells from human osteoporotic patients. Notably, transplantation of Nfic-overexpressing BMSCs stimulates osteoblast differentiation and new bone formation, but inhibits adipocyte differentiation by suppressing peroxisome proliferator-activated receptor gamma expression in Nfic(-/-) mice showing an age-related osteoporosis-like phenotype. Finally, NFI-C directly regulates Osterix expression but acts downstream of the bone morphogenetic protein-2-Runx2 pathway. These results suggest that NFI-C acts as a transcriptional switch in cell fate determination between osteoblast and adipocyte differentiation in BMSCs. Therefore, regulation of NFI-C expression in BMSCs could be a novel therapeutic approach for treating age-related osteoporosis.


Assuntos
Fatores de Transcrição NFI/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Fatores de Transcrição/biossíntese , Idoso , Animais , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Transcrição NFI/genética , Osteogênese/fisiologia , Fator de Transcrição Sp7 , Transfecção
18.
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
19.
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
20.
BMC Genomics ; 15: 231, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24661679

RESUMO

BACKGROUND: Lung maturation is a late fetal developmental event in both mice and humans. Because of this, lung immaturity is a serious problem in premature infants. Disruption of genes for either the glucocorticoid receptor (Nr3c1) or the NFIB transcription factors results in perinatal lethality due to lung immaturity. In both knockouts, the phenotype includes excess cell proliferation, failure of saccularization and reduced expression of markers of epithelial differentiation. This similarity suggests that the two genes may co-regulate a specific set of genes essential for lung maturation. RESULTS: We analyzed the roles of these two transcription factors in regulating transcription using ChIP-seq data for NFIB, and RNA expression data and motif analysis for both. Our new ChIP-seq data for NFIB in lung at E16.5 shows that NFIB binds to a NFI motif. This motif is over-represented in the promoters of genes that are under-expressed in Nfib-KO mice at E18.5, suggesting an activator role for NFIB. Using available microarray data from Nr3c1-KO mice, we further identified 52 genes that are under-expressed in both Nfib and Nr3c1 knockouts, an overlap which is 13.1 times larger than what would be expected by chance. Finally, we looked for enrichment of 738 recently published transcription factor motifs in the promoters of these putative target genes and found that the NFIB and glucocorticoid receptor motifs were among the most enriched, suggesting that a subset of these genes may be directly activated by Nfib and Nr3c1. CONCLUSIONS: Our data provide the first evidence for Nfib and Nr3c1 co-regulating genes related to lung maturation. They also establish that the in vivo DNA-binding specificity of NFIB is the same as previously seen in vitro, and highly similar to that of the other NFI-family members NFIA, NFIC and NFIX.


Assuntos
Pulmão/metabolismo , Fatores de Transcrição NFI/genética , Receptores de Glucocorticoides/genética , Animais , Sequência de Bases , Imunoprecipitação da Cromatina , DNA/metabolismo , Desenvolvimento Embrionário/genética , Humanos , Pulmão/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Fatores de Transcrição NFI/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Receptores de Glucocorticoides/metabolismo , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA