Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brief Funct Genomics ; 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38183207

RESUMEN

Metastatic melanoma originates from melanocytes of the skin. Melanoma metastasis results in poor treatment prognosis for patients and is associated with epigenetic and transcriptional changes that reflect the developmental program of melanocyte differentiation from neural crest stem cells. Several studies have explored melanoma transcriptional heterogeneity using microarray, bulk and single-cell RNA-sequencing technologies to derive data-driven models of the transcriptional-state change which occurs during melanoma progression. No study has systematically examined how different models of melanoma progression derived from different data types, technologies and biological conditions compare. Here, we perform a cross-sectional study to identify averaging effects of bulk-based studies that mask and distort apparent melanoma transcriptional heterogeneity; we describe new transcriptionally distinct melanoma cell states, identify differential co-expression of genes between studies and examine the effects of predicted drug susceptibilities of different cell states between studies. Importantly, we observe considerable variability in drug-target gene expression between studies, indicating potential transcriptional plasticity of melanoma to down-regulate these drug targets and thereby circumvent treatment. Overall, observed differences in gene co-expression and predicted drug susceptibility between studies suggest bulk-based transcriptional measurements do not reliably gauge heterogeneity and that melanoma transcriptional plasticity is greater than described when studies are considered in isolation.

2.
Cells ; 11(15)2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35954220

RESUMEN

Nuclear factor one X (NFIX) is a transcription factor required for normal ependymal development. Constitutive loss of Nfix in mice (Nfix-/-) is associated with hydrocephalus and sloughing of the dorsal ependyma within the lateral ventricles. Previous studies have implicated NFIX in the transcriptional regulation of genes encoding for factors essential to ependymal development. However, the cellular and molecular mechanisms underpinning hydrocephalus in Nfix-/- mice are unknown. To investigate the role of NFIX in hydrocephalus, we examined ependymal cells in brains from postnatal Nfix-/- and control (Nfix+/+) mice using a combination of confocal and electron microscopy. This revealed that the ependymal cells in Nfix-/- mice exhibited abnormal cilia structure and disrupted localisation of adhesion proteins. Furthermore, we modelled ependymal cell adhesion using epithelial cell culture and revealed changes in extracellular matrix and adherens junction gene expression following knockdown of NFIX. Finally, the ablation of Nfix from ependymal cells in the adult brain using a conditional approach culminated in enlarged ventricles, sloughing of ependymal cells from the lateral ventricles and abnormal localisation of adhesion proteins, which are phenotypes observed during development. Collectively, these data demonstrate a pivotal role for NFIX in the regulation of cell adhesion within ependymal cells of the lateral ventricles.


Asunto(s)
Epéndimo , Hidrocefalia , Factores de Transcripción NFI , Animales , Fenómenos Fisiológicos Celulares , Hidrocefalia/genética , Ventrículos Laterales , Ratones , Factores de Transcripción NFI/genética , Neuroglía
3.
BMC Res Notes ; 14(1): 269, 2021 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-34256843

RESUMEN

OBJECTIVE: Sulfation is an essential physiological process that regulates the function of a wide array of molecules involved in brain development. We have previously shown expression levels for the sulfate transporter Slc13a4 to be elevated during postnatal development, and that sulfate accumulation in the brains of Slc13a4+/- mice is reduced, suggesting a role for this transporter during this critical window of brain development. In order to understand the pathways regulated by cellular sulfation within the brain, we performed a bulk RNA-sequencing analysis of the forebrain of postnatal day 20 (P20) Slc13a4 heterozygous mice and wild-type litter mate controls. DATA DESCRIPTION: We performed an RNA transcriptomic based sequencing screen on the whole forebrain from Slc13a4+/- and Slc13a4+/+mice at P20. Differential expression analysis revealed 90 differentially regulated genes in the forebrain of Slc13a4+/- mice (a p-value of 0.1 was considered as significant). Of these, 55 were upregulated, and 35 were downregulated in the forebrain of heterozygous mice. Moreover, when we stratified further with a ± 1.2 fold-change, we observed 38 upregulated, and 16 downregulated genes in the forebrain of heterozygous mice. This resource provides a useful tool to interrogate which pathways may require elevated sulfate levels to drive normal postnatal development of the brain.


Asunto(s)
Simportadores , Animales , Perfilación de la Expresión Génica , Ratones , Prosencéfalo/metabolismo , Transportadores de Sulfato , Simportadores/genética , Transcriptoma
4.
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
5.
Biol Open ; 9(7)2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32661132

RESUMEN

Sulfate is a key anion required for a range of physiological functions within the brain. These include sulfonation of extracellular proteoglycans to facilitate local growth factor binding and to regulate the shape of morphogen gradients during development. We have previously shown that mice lacking one allele of the sulfate transporter Slc13a4 exhibit reduced sulfate transport into the brain, deficits in social behaviour, reduced performance in learning and memory tasks, and abnormal neurogenesis within the ventricular/subventricular zone lining the lateral ventricles. However, whether these mice have deficits in hippocampal neurogenesis was not addressed. Here, we demonstrate that adult Slc13a4+/- mice have increased neurogenesis within the subgranular zone (SGZ) of the hippocampal dentate gyrus, with elevated numbers of neural progenitor cells and intermediate progenitors. In contrast, by 12 months of age there were reduced numbers of neural stem cells in the SGZ of heterozygous mice. Importantly, we did not observe any changes in proliferation when we isolated and cultured progenitors in vitro in neurosphere assays, suggestive of a cell-extrinsic requirement for sulfate in regulating hippocampal neurogenesis. Collectively, these data demonstrate a requirement for sulfate transport during postnatal brain development to ensure normal adult hippocampal neurogenesis.


Asunto(s)
Hipocampo/fisiología , Neurogénesis , Sulfatos/metabolismo , Animales , Biomarcadores , Diferenciación Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Hipocampo/crecimiento & desarrollo , Ratones , Ratones Noqueados , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Piramidales/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Simportadores/genética , Simportadores/metabolismo
6.
Gene Expr Patterns ; 35: 119098, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-32068188

RESUMEN

The Nuclear factor I proteins comprise a family of transcription factors that are expressed in many developing and mature cell populations, including within the central nervous system. Within the embryonic mouse spinal cord, NFIA and NFIB are expressed by neural progenitor cells lining the central canal, where they act to promote astrocytic and oligodendrocytic lineage specification. Cells lining the mature spinal cord central canal retain characteristics of neural progenitor cells, but the expression of NFIA and NFIB within the mature spinal cord at a cell-type-specific level remains undefined. Here, we investigated where these two transcription factors are expressed within the adult mouse spinal cord. We reveal that both factors are expressed in similar cohorts of mature cells, including ependymal cells, interneurons and motor neurons. We also show robust and widespread expression of NFIA and NFIB within nestin-expressing cells following injury to the spinal cord. Collectively, these data provide a basis to further define what functional role(s) NFIA and NFIB play within the adult spinal cord.


Asunto(s)
Factores de Transcripción NFI/genética , Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/metabolismo , Animales , Epéndimo/citología , Epéndimo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Médula Espinal/citología
7.
Genes Brain Behav ; 19(4): e12637, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31909872

RESUMEN

Sotos syndrome is a developmental disorder characterized by a suite of clinical features. In children, the three cardinal features of Sotos syndrome are a characteristic facial appearance, learning disability and overgrowth (height and/or head circumference > 2 SDs above average). These features are also evident in adults with this syndrome. Over 90% of Sotos syndrome patients are haploinsufficient for the gene encoding nuclear receptor-binding Su(var)3-9, Enhancer-of-zesteand Trithorax domain-containing protein 1 (NSD1). NSD1 is a histone methyltransferase that catalyzes the methylation of lysine residue 36 on histone H3. However, although the symptomology of Sotos syndrome is well established, many aspects of NSD1 biology remain unknown. Here, we assessed the expression of Nsd1 within the mouse brain, and showed a predominantly neuronal pattern of expression for this histone-modifying factor. We also generated a mouse strain lacking one allele of Nsd1 and analyzed morphological and behavioral characteristics in these mice, showing behavioral characteristics reminiscent of some of the deficits seen in Sotos syndrome patients.


Asunto(s)
Corteza Cerebral/patología , N-Metiltransferasa de Histona-Lisina/genética , Síndrome de Sotos/genética , Animales , Corteza Cerebral/metabolismo , Femenino , Heterocigoto , N-Metiltransferasa de Histona-Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Síndrome de Sotos/patología
8.
Cerebellum ; 19(1): 89-101, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31838646

RESUMEN

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.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Factores de Transcripción NFI/metabolismo , Animales , Animales Recién Nacidos , Cerebelo/citología , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/genética , Neurogénesis/fisiología , Embarazo
9.
Cereb Cortex ; 29(8): 3590-3604, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-30272140

RESUMEN

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.


Asunto(s)
Movimiento Celular/genética , Giro Dentado/citología , Ventrículos Laterales/citología , Factores de Transcripción NFI/genética , Células-Madre Neurales/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Ratones , Células-Madre Neurales/citología , Neurogénesis/genética , Receptores del Factor Natriurético Atrial/genética
10.
Brain Struct Funct ; 224(2): 811-827, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30511336

RESUMEN

Cerebellar granule neurons are the most numerous neuronal subtype in the central nervous system. Within the developing cerebellum, these neurons are derived from a population of progenitor cells found within the external granule layer of the cerebellar anlage, namely the cerebellar granule neuron precursors (GNPs). The timely proliferation and differentiation of these precursor cells, which, in rodents occurs predominantly in the postnatal period, is tightly controlled to ensure the normal morphogenesis of the cerebellum. Despite this, our understanding of the factors mediating how GNP differentiation is controlled remains limited. Here, we reveal that the transcription factor nuclear factor I X (NFIX) plays an important role in this process. Mice lacking Nfix exhibit reduced numbers of GNPs during early postnatal development, but elevated numbers of these cells at postnatal day 15. Moreover, Nfix-/- GNPs exhibit increased proliferation when cultured in vitro, suggestive of a role for NFIX in promoting GNP differentiation. At a mechanistic level, profiling analyses using both ChIP-seq and RNA-seq identified the actin-associated factor intersectin 1 as a downstream target of NFIX during cerebellar development. In support of this, mice lacking intersectin 1 also displayed delayed GNP differentiation. Collectively, these findings highlight a key role for NFIX and intersectin 1 in the regulation of cerebellar development.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proliferación Celular/fisiología , Cerebelo/citología , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Neuronas/citología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones Noqueados , Factores de Transcripción NFI/genética , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo
12.
Development ; 145(3)2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29437824

RESUMEN

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.


Asunto(s)
Hipocampo/citología , Hipocampo/metabolismo , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Supervivencia Celular , Proteína Doblecortina , Femenino , Regulación del Desarrollo de la Expresión Génica , Hipocampo/crecimiento & desarrollo , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Nicho de Células Madre/genética , Nicho de Células Madre/fisiología , Regulación hacia Arriba
13.
Neural Dev ; 13(1): 2, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29452604

RESUMEN

BACKGROUND: Radial glial stem cells within the developing nervous system generate a variety of post-mitotic cells, including neurons and glial cells, as well as the specialised multi-ciliated cells that line the walls of the ventricular system, the ependymal cells. Ependymal cells separate the brain parenchyma from the cerebrospinal fluid and mediate osmotic regulation, the flow of cerebrospinal fluid, and the subsequent dispersion of signalling molecules via the co-ordinated beating of their cilia. Deficits to ependymal cell development and function have been implicated in the formation of hydrocephalus, but the transcriptional mechanisms underpinning ependymal development remain poorly characterised. FINDINGS: Here, we demonstrate that the transcription factor nuclear factor IX (NFIX) plays a central role in the development of the ependymal cell layer of the lateral ventricles. Expression of ependymal cell-specific markers is delayed in the absence of Nfix. Moreover, Nfix-deficient mice exhibit aberrant ependymal cell morphology at postnatal day 15, culminating in abnormal thickening and intermittent loss of this cell layer. Finally, we reveal Foxj1, a key factor promoting ependymal cell maturation, as a target for NFIX-mediated transcriptional activation. CONCLUSIONS: Collectively, our data indicate that ependymal cell development is reliant, at least in part, on NFIX expression, further implicating this transcription factor as a mediator of multiple aspects of radial glial biology during corticogenesis.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/crecimiento & desarrollo , Epéndimo/citología , Neuroglía/metabolismo , Factores de Transcripción/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Cadherinas/metabolismo , Inmunoprecipitación de Cromatina , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción/genética
14.
J Comp Neurol ; 525(11): 2465-2483, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28295292

RESUMEN

The nuclear factor I (NFI) family of transcription factors plays an important role in the development of the cerebral cortex in humans and mice. Disruption of nuclear factor IA (NFIA), nuclear factor IB (NFIB), or nuclear factor IX (NFIX) results in abnormal development of the corpus callosum, lateral ventricles, and hippocampus. However, the expression or function of these genes has not been examined in detail in the adult brain, and the cell type-specific expression of NFIA, NFIB, and NFIX is currently unknown. Here, we demonstrate that the expression of each NFI protein shows a distinct laminar pattern in the adult mouse neocortex and that their cell type-specific expression differs depending on the family member. NFIA expression was more frequently observed in astrocytes and oligodendroglia, whereas NFIB expression was predominantly localized to astrocytes and neurons. NFIX expression was most commonly observed in neurons. The NFI proteins were equally distributed within microglia, and the ependymal cells lining the ventricles of the brain expressed all three proteins. In the hippocampus, the NFI proteins were expressed during all stages of neural stem cell differentiation in the dentate gyrus, with higher expression intensity in neuroblast cells as compared to quiescent stem cells and mature granule neurons. These findings suggest that the NFI proteins may play distinct roles in cell lineage specification or maintenance, and establish the basis for further investigation of their function in the adult brain and their emerging role in disease.


Asunto(s)
Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción NFI/biosíntesis , Neuroglía/metabolismo , Neuronas/metabolismo , Factores de Edad , Animales , Diferenciación Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Factores de Transcripción NFI/genética
15.
EBioMedicine ; 16: 63-75, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28119061

RESUMEN

While invasion and metastasis of tumour cells are the principle factor responsible for cancer related deaths, the mechanisms governing the process remain poorly defined. Moreover, phenotypic divergence of sub-populations of tumour cells is known to underpin alternative behaviors linked to tumour progression such as proliferation, survival and invasion. In the context of melanoma, heterogeneity between two transcription factors, BRN2 and MITF, has been associated with phenotypic switching between predominantly invasive and proliferative behaviors respectively. Epigenetic changes, in response to external cues, have been proposed to underpin this process, however the mechanism by which the phenotypic switch occurs is unclear. Here we report the identification of the NFIB transcription factor as a novel downstream effector of BRN2 function in melanoma cells linked to the migratory and invasive characteristics of these cells. Furthermore, the function of NFIB appears to drive an invasive phenotype through an epigenetic mechanism achieved via the upregulation of the polycomb group protein EZH2. A notable target of NFIB mediated up-regulation of EZH2 is decreased MITF expression, which further promotes a less proliferative, more invasive phenotype. Together our data reveal that NFIB has the ability to promote dynamic changes in the chromatin state of melanoma cells to facilitate migration, invasion and metastasis.


Asunto(s)
Movimiento Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas de Homeodominio/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factores de Transcripción NFI/genética , Factores del Dominio POU/genética , Animales , Western Blotting , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones Endogámicos BALB C , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/metabolismo , Microscopía Fluorescente , Factores de Transcripción NFI/metabolismo , Invasividad Neoplásica , Factores del Dominio POU/metabolismo , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
16.
Brain Struct Funct ; 222(5): 2251-2270, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27878595

RESUMEN

Transcription factors from the nuclear factor one (NFI) family have been shown to play a central role in regulating neural progenitor cell differentiation within the embryonic and post-natal brain. NFIA and NFIB, for instance, promote the differentiation and functional maturation of granule neurons within the cerebellum. Mice lacking Nfix exhibit delays in the development of neuronal and glial lineages within the cerebellum, but the cell-type-specific expression of this transcription factor remains undefined. Here, we examined the expression of NFIX, together with various cell-type-specific markers, within the developing and adult cerebellum using both chromogenic immunohistochemistry and co-immunofluorescence labelling and confocal microscopy. In embryos, NFIX was expressed by progenitor cells within the rhombic lip and ventricular zone. After birth, progenitor cells within the external granule layer, as well as migrating and mature granule neurons, expressed NFIX. Within the adult cerebellum, NFIX displayed a broad expression profile, and was evident within granule cells, Bergmann glia, and interneurons, but not within Purkinje neurons. Furthermore, transcriptomic profiling of cerebellar granule neuron progenitor cells showed that multiple splice variants of Nfix are expressed within this germinal zone of the post-natal brain. Collectively, these data suggest that NFIX plays a role in regulating progenitor cell biology within the embryonic and post-natal cerebellum, as well as an ongoing role within multiple neuronal and glial populations within the adult cerebellum.


Asunto(s)
Diferenciación Celular/fisiología , Cerebelo/citología , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Envejecimiento , Animales , Astrocitos/metabolismo , Cerebelo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones Endogámicos C57BL , Neurogénesis/fisiología , Neuronas/metabolismo
17.
J Mol Histol ; 48(1): 53-61, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27803996

RESUMEN

Endothelial cells form a critical component of the coronary vasculature, yet the factors regulating their development remain poorly defined. Here we reveal a novel role for the transmembrane protein CRIM1 in mediating cardiac endothelial cell development. In the absence of Crim1 in vivo, the coronary vasculature is malformed, the number of endothelial cells reduced, and the canonical BMP pathway dysregulated. Moreover, we reveal that CRIM1 can bind IGFs, and regulate IGF signalling within endothelial cells. Finally, loss of CRIM1 from human cardiac endothelial cells results in misregulation of endothelial genes, predicted by pathway analysis to be involved in an increased inflammatory response and cytolysis, reminiscent of endothelial cell dysfunction in cardiovascular disease pathogenesis. Collectively, these findings implicate CRIM1 in endothelial cell development and homeostasis in the coronary vasculature.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Vasos Coronarios/citología , Células Endoteliales/metabolismo , Homeostasis , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Supervivencia Celular/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Mutación , Transducción de Señal
18.
Development ; 143(24): 4620-4630, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27965439

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Hipocampo/embriología , Factores de Transcripción NFI/genética , Células-Madre Neurales/citología , Neurogénesis/genética , Animales , Proteínas de Ciclo Celular/genética , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Neurogénesis/fisiología , Neuronas/citología , Regiones Promotoras Genéticas/genética , Transcripción Genética , Activación Transcripcional/genética
19.
Sci Rep ; 6: 25783, 2016 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-27181636

RESUMEN

Within the adult mammalian brain, neurogenesis persists within two main discrete locations, the subventricular zone lining the lateral ventricles, and the hippocampal dentate gyrus. Neurogenesis within the adult dentate gyrus contributes to learning and memory, and deficiencies in neurogenesis have been linked to cognitive decline. Neural stem cells within the adult dentate gyrus reside within the subgranular zone (SGZ), and proteins intrinsic to stem cells, and factors within the niche microenvironment, are critical determinants for development and maintenance of this structure. Our understanding of the repertoire of these factors, however, remains limited. The deubiquitylating enzyme USP9X has recently emerged as a mediator of neural stem cell identity. Furthermore, mice lacking Usp9x exhibit a striking reduction in the overall size of the adult dentate gyrus. Here we reveal that the development of the postnatal SGZ is abnormal in mice lacking Usp9x. Usp9x conditional knockout mice exhibit a smaller hippocampus and shortened dentate gyrus blades from as early as P7. Moreover, the analysis of cellular populations within the dentate gyrus revealed reduced stem cell, neuroblast and neuronal numbers and abnormal neuroblast morphology. Collectively, these findings highlight the critical role played by USP9X in the normal morphological development of the postnatal dentate gyrus.


Asunto(s)
Giro Dentado/crecimiento & desarrollo , Giro Dentado/metabolismo , Endopeptidasas/deficiencia , Animales , Animales Recién Nacidos , Recuento de Células , Diferenciación Celular , Giro Dentado/citología , Endopeptidasas/metabolismo , Femenino , Integrasas/metabolismo , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Tamaño de los Órganos , Ubiquitina Tiolesterasa
20.
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
DETALLE DE LA BÚSQUEDA
...