Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Genes Dev ; 27(16): 1769-86, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23964093

RESUMEN

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.


Asunto(s)
Epigénesis Genética , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Proteína Morfogenética Ósea 4/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Factores de Transcripción NFI/genética , Células-Madre Neurales/efectos de los fármacos , Unión Proteica
2.
Am J Med Genet C Semin Med Genet ; 181(4): 611-626, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31730271

RESUMEN

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.


Asunto(s)
Crecimiento/genética , Mutación , Factores de Transcripción NFI/genética , Anomalías Múltiples/genética , Animales , Enfermedades del Desarrollo Óseo/genética , Anomalías Craneofaciales/genética , Duplicación de Gen , Trastornos del Crecimiento/genética , Humanos , Ratones , Displasia Septo-Óptica/genética , Síndrome
3.
Cereb Cortex ; 25(10): 3758-78, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25331604

RESUMEN

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.


Asunto(s)
Movimiento Celular , Proliferación Celular , Ventrículos Laterales/embriología , Factores de Transcripción NFI/fisiología , Células-Madre Neurales/fisiología , Neurogénesis , Animales , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Interneuronas/fisiología , Ventrículos Laterales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Neuroglía/fisiología , Bulbo Olfatorio/embriología , Bulbo Olfatorio/metabolismo , Nicho de Células Madre
4.
Dev Dyn ; 244(3): 227-38, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25156673

RESUMEN

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.


Asunto(s)
Células Madre Adultas/metabolismo , Diferenciación Celular/fisiología , Factores de Transcripción NFI/metabolismo , Células Madre Adultas/citología , Animales , Supervivencia Celular/fisiología , Humanos , Ratones , Ratones Mutantes , Factores de Transcripción NFI/genética
5.
Cereb Cortex ; 24(1): 261-79, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23042739

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Hipocampo/embriología , Factores de Transcripción NFI/fisiología , Células-Madre Neurales/fisiología , Animales , Recuento de Células , Colorantes , Biología Computacional , Giro Dentado/embriología , Giro Dentado/crecimiento & desarrollo , Giro Dentado/fisiología , Ensayo de Cambio de Movilidad Electroforética , Electroporación , Femenino , Hematoxilina , Hipocampo/citología , Hipocampo/metabolismo , Inmunohistoquímica , Hibridación in Situ , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Factores de Transcripción NFI/genética , Células-Madre Neurales/metabolismo , Adhesión en Parafina , Embarazo , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
BMC Res Notes ; 13(1): 437, 2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938475

RESUMEN

OBJECTIVE: Nuclear Factor One X (NFIX) is a transcription factor expressed by neural stem cells within the developing mouse brain and spinal cord. In order to characterise the pathways by which NFIX may regulate neural stem cell biology within the developing mouse spinal cord, we performed an microarray-based transcriptomic analysis of the spinal cord of embryonic day (E)14.5 Nfix-/- mice in comparison to wild-type controls. DATA DESCRIPTION: Using microarray and differential gene expression analyses, we were able to identify differentially expressed genes in the spinal cords of E14.5 Nfix-/- mice compared to wild-type controls. We performed microarray-based sequencing on spinal cords from n = 3 E14.5 Nfix-/- mice and n = 3 E14.5 Nfix+/+ mice. Differential gene expression analysis, using a false discovery rate (FDR) p-value of p < 0.05, and a fold change cut-off for differential expression of > ± 1.5, revealed 1351 differentially regulated genes in the spinal cord of Nfix-/- mice. Of these, 828 were upregulated, and 523 were downregulated. This resource provides a tool to interrogate the role of this transcription factor in spinal cord development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción NFI , Animales , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFI/genética , Médula Espinal
7.
Cancer Lett ; 410: 124-138, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28962832

RESUMEN

The nuclear factor I (NFI) transcription factors play important roles during normal development and have been associated with developmental abnormalities in humans. All four family members, NFIA, NFIB, NFIC and NFIX, have a homologous DNA binding domain and function by regulating cell proliferation and differentiation via the transcriptional control of their target genes. More recently, NFI genes have also been implicated in cancer based on genomic analyses and studies of animal models in a variety of tumours across multiple organ systems. However, the association between their functions in development and in cancer is not well described. In this review, we summarise the evidence suggesting a converging role for the NFI genes in development and cancer. Our review includes all cancer types in which the NFI genes are implicated, focusing predominantly on studies demonstrating their oncogenic or tumour-suppressive potential. We conclude by presenting the challenges impeding our understanding of NFI function in cancer biology, and demonstrate how a developmental perspective may contribute towards overcoming such hurdles.


Asunto(s)
Factores de Transcripción NFI/metabolismo , Neoplasias/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Factores de Transcripción NFI/genética , Neoplasias/genética , Neoplasias/patología , Transducción de Señal , Transcripción Genética
8.
Brain Res ; 1616: 71-87, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25960350

RESUMEN

Nuclear factor one X (NFIX) has been shown to play a pivotal role during the development of many regions of the brain, including the neocortex, the hippocampus and the cerebellum. Mechanistically, NFIX has been shown to promote neural stem cell differentiation through the activation of astrocyte-specific genes and via the repression of genes central to progenitor cell self-renewal. Interestingly, mice lacking Nfix also exhibit other phenotypes with respect to development of the central nervous system, and whose underlying causes have yet to be determined. Here we examine one of the phenotypes displayed by Nfix(-/-) mice, namely hydrocephalus. Through the examination of embryonic and postnatal Nfix(-/-) mice we reveal that hydrocephalus is first seen at around postnatal day (P) 10 in mice lacking Nfix, and is fully penetrant by P20. Furthermore, we examined the subcommissural organ (SCO), the Sylvian aqueduct and the ependymal layer of the lateral ventricles, regions that when malformed and functionally perturbed have previously been implicated in the development of hydrocephalus. SOX3 is a factor known to regulate SCO development. Although we revealed that NFIX could repress Sox3-promoter-driven transcriptional activity in vitro, SOX3 expression within the SCO was normal within Nfix(-/-) mice, and Nfix mutant mice showed no abnormalities in the structure or function of the SCO. Moreover, these mutant mice exhibited no overt blockage of the Sylvian aqueduct. However, the ependymal layer of the lateral ventricles was frequently absent in Nfix(-/-) mice, suggesting that this phenotype may underlie the development of hydrocephalus within these knockout mice.


Asunto(s)
Epéndimo/patología , Regulación del Desarrollo de la Expresión Génica/genética , Hidrocefalia/patología , Ventrículos Laterales/patología , Factores de Transcripción NFI/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Biología Computacional , Modelos Animales de Enfermedad , Embrión de Mamíferos , Epéndimo/embriología , Epéndimo/crecimiento & desarrollo , Hidrocefalia/genética , Ventrículos Laterales/embriología , Ventrículos Laterales/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción NFI/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda