Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Development ; 149(14)2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35861233

RESUMEN

The transcription factor SOX2 is a vital regulator of stem cell activity in various developing and adult tissues. Mounting evidence has demonstrated the importance of SOX2 in regulating the induction and maintenance of stemness as well as in controlling cell proliferation, lineage decisions and differentiation. Recent studies have revealed that the ability of SOX2 to regulate these stem cell features involves its function as a pioneer factor, with the capacity to target nucleosomal DNA, modulate chromatin accessibility and prepare silent genes for subsequent activation. Moreover, although SOX2 binds to similar DNA motifs in different stem cells, its multifaceted and cell type-specific functions are reliant on context-dependent features. These cell type-specific properties include variations in partner factor availability and SOX2 protein expression levels. In this Primer, we discuss recent findings that have increased our understanding of how SOX2 executes its versatile functions as a master regulator of stem cell activities.


Asunto(s)
Nucleosomas , Factores de Transcripción SOXB1 , Diferenciación Celular/genética , Proliferación Celular/genética , Cromatina , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(34): 16997-17006, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31375625

RESUMEN

Despite the discovery of the oxygen-sensitive regulation of HIFα by the von Hippel-Lindau (VHL) protein, the mechanisms underlying the complex genotype/phenotype correlations in VHL disease remain unknown. Some germline VHL mutations cause familial pheochromocytoma and encode proteins that preserve their ability to down-regulate HIFα. While type 1, 2A, and 2B VHL mutants are defective in regulating HIFα, type 2C mutants encode proteins that preserve their ability to down-regulate HIFα. Here, we identified an oxygen-sensitive function of VHL that is abolished by VHL type 2C mutations. We found that BIM-EL, a proapoptotic BH3-only protein, is hydroxylated by EglN3 and subsequently bound by VHL. VHL mutants fail to bind hydroxylated BIM-EL, regardless of whether they have the ability to bind hydroxylated HIFα or not. VHL binding inhibits BIM-EL phosphorylation by extracellular signal-related kinase (ERK) on serine 69. This causes BIM-EL to escape from proteasomal degradation, allowing it to enhance EglN3-induced apoptosis. BIM-EL was rapidly degraded in cells lacking wild-type VHL or in which EglN3 was inactivated genetically or by lack of oxygen, leading to enhanced cell survival and chemotherapy resistance. Combination therapy using ERK inhibitors, however, resensitizes VHL- and EglN3-deficient cells that are otherwise cisplatin-resistant.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Proteína 11 Similar a Bcl2/metabolismo , Resistencia a Antineoplásicos/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Mutación , Feocromocitoma , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Neoplasias de las Glándulas Suprarrenales/tratamiento farmacológico , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/metabolismo , Neoplasias de las Glándulas Suprarrenales/patología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína 11 Similar a Bcl2/genética , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Hidroxilación/efectos de los fármacos , Hidroxilación/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Células PC12 , Feocromocitoma/tratamiento farmacológico , Feocromocitoma/metabolismo , Feocromocitoma/patología , Proteolisis/efectos de los fármacos , Ratas , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
3.
PLoS Genet ; 14(2): e1007224, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29432416

RESUMEN

Stem cells are defined by their capacities to self-renew and generate progeny of multiple lineages. The transcription factor SOX2 has key roles in the regulation of stem cell characteristics, but whether SOX2 achieves these functions through similar mechanisms in distinct stem cell populations is not known. To address this question, we performed RNA-seq and SOX2 ChIP-seq on embryonic mouse cortex, spinal cord, stomach and lung/esophagus. We demonstrate that, although SOX2 binds a similar motif in the different cell types, its target regions are primarily cell-type-specific and enriched for the distinct binding motifs of appropriately expressed interacting co-factors. Furthermore, cell-type-specific SOX2 binding in endodermal and neural cells is most often found around genes specifically expressed in the corresponding tissue. Consistent with this, we demonstrate that SOX2 target regions can act as cis-regulatory modules capable of directing reporter expression to appropriate tissues in a zebrafish reporter assay. In contrast, SOX2 binding sites found in both endodermal and neural tissues are associated with genes regulating general stem cell features, such as proliferation. Notably, we provide evidence that SOX2 regulates proliferation through conserved mechanisms and target genes in both germ layers examined. Together, these findings demonstrate how SOX2 simultaneously regulates cell-type-specific, as well as core transcriptional programs in neural and endodermal stem cells.


Asunto(s)
Sistema Nervioso Central/embriología , Endodermo/citología , Endodermo/embriología , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/fisiología , Células-Madre Neurales/fisiología , Organogénesis/genética , Factores de Transcripción SOXB1/fisiología , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Sistema Nervioso Central/citología , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Transgénicos , Células-Madre Neurales/citología , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción SOXB1/genética
4.
EMBO Rep ; 19(11)2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30166336

RESUMEN

SOX transcription factors have important roles during astrocyte and oligodendrocyte development, but how glial genes are specified and activated in a sub-lineage-specific fashion remains unknown. Here, we define glial-specific gene expression in the developing spinal cord using single-cell RNA-sequencing. Moreover, by ChIP-seq analyses we show that these glial gene sets are extensively preselected already in multipotent neural precursor cells through prebinding by SOX3. In the subsequent lineage-restricted glial precursor cells, astrocyte genes become additionally targeted by SOX9 at DNA regions strongly enriched for Nfi binding motifs. Oligodendrocyte genes instead are prebound by SOX9 only, at sites which during oligodendrocyte maturation are targeted by SOX10. Interestingly, reporter gene assays and functional studies in the spinal cord reveal that SOX3 binding represses the synergistic activation of astrocyte genes by SOX9 and NFIA, whereas oligodendrocyte genes are activated in a combinatorial manner by SOX9 and SOX10. These genome-wide studies demonstrate how sequentially expressed SOX proteins act on lineage-specific regulatory DNA elements to coordinate glial gene expression both in a temporal and in a sub-lineage-specific fashion.


Asunto(s)
Astrocitos/fisiología , Oligodendroglía/fisiología , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXB1/genética , Médula Espinal/citología , Animales , Diferenciación Celular/genética , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Ratones , Células-Madre Neurales , Neuroglía/citología , Neuroglía/fisiología , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXB1/metabolismo , Médula Espinal/crecimiento & desarrollo
5.
Genes Dev ; 25(23): 2453-64, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22085726

RESUMEN

Pluripotent embryonic stem (ES) cells can generate all cell types, but how cell lineages are initially specified and maintained during development remains largely unknown. Different classes of Sox transcription factors are expressed during neurogenesis and have been assigned important roles from early lineage specification to neuronal differentiation. Here we characterize the genome-wide binding for Sox2, Sox3, and Sox11, which have vital functions in ES cells, neural precursor cells (NPCs), and maturing neurons, respectively. The data demonstrate that Sox factor binding depends on developmental stage-specific constraints and reveal a remarkable sequential binding of Sox proteins to a common set of neural genes. Interestingly, in ES cells, Sox2 preselects for neural lineage-specific genes destined to be bound and activated by Sox3 in NPCs. In NPCs, Sox3 binds genes that are later bound and activated by Sox11 in differentiating neurons. Genes prebound by Sox proteins are associated with a bivalent chromatin signature, which is resolved into a permissive monovalent state upon binding of activating Sox factors. These data indicate that a single key transcription factor family acts sequentially to coordinate neural gene expression from the early lineage specification in pluripotent cells to later stages of neuronal development.


Asunto(s)
Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Neuronas/citología , Factores de Transcripción SOX/genética , Animales , Línea Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genoma , Ratones , Neurogénesis/genética , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOX/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXC/genética , Regulación hacia Arriba
6.
Proc Natl Acad Sci U S A ; 110(18): 7330-5, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589857

RESUMEN

The reiterative deployment of a small cadre of morphogen signals underlies patterning and growth of most tissues during embyogenesis, but how such inductive events result in tissue-specific responses remains poorly understood. By characterizing cis-regulatory modules (CRMs) associated with genes regulated by Sonic hedgehog (Shh), retinoids, or bone morphogenetic proteins in the CNS, we provide evidence that the neural-specific interpretation of morphogen signaling reflects a direct integration of these pathways with SoxB1 proteins at the CRM level. Moreover, expression of SoxB1 proteins in the limb bud confers on mesodermal cells the potential to activate neural-specific target genes upon Shh, retinoid, or bone morphogenetic protein signaling, and the collocation of binding sites for SoxB1 and morphogen-mediatory transcription factors in CRMs faithfully predicts neural-specific gene activity. Thus, an unexpectedly simple transcriptional paradigm appears to conceptually explain the neural-specific interpretation of pleiotropic signaling during vertebrate development. Importantly, genes induced in a SoxB1-dependent manner appear to constitute repressive gene regulatory networks that are directly interlinked at the CRM level to constrain the regional expression of patterning genes. Accordingly, not only does the topology of SoxB1-driven gene regulatory networks provide a tissue-specific mode of gene activation, but it also determines the spatial expression pattern of target genes within the developing neural tube.


Asunto(s)
Redes Reguladoras de Genes/genética , Neuronas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/genética , Animales , Sitios de Unión , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/farmacología , Pollos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Genoma/genética , Proteínas Hedgehog/metabolismo , Esbozos de los Miembros/efectos de los fármacos , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retinoides/farmacología , Transducción de Señal/efectos de los fármacos
7.
Stem Cells ; 32(9): 2539-49, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24807147

RESUMEN

Degeneration of central nervous system tissue commonly occurs during neuroinflammatory conditions, such as multiple sclerosis and neurotrauma. During such conditions, neural stem/progenitor cell (NPC) populations have been suggested to provide new cells to degenerated areas. In the normal brain, NPCs from the subventricular zone generate neurons that settle in the olfactory bulb or striatum. However, during neuroinflammatory conditions NPCs migrate toward the site of injury to form oligodendrocytes and astrocytes, whereas newly formed neurons are less abundant. Thus, the specific NPC lineage fate decisions appear to respond to signals from the local environment. The instructive signals from inflammation have been suggested to rely on excessive levels of the free radical nitric oxide (NO), which is an essential component of the innate immune response, as NO promotes neuronal to glial cell fate conversion of differentiating rat NPCs in vitro. Here, we demonstrate that the NO-induced neuronal to glial fate conversion is dependent on the transcription factor neuron-restrictive silencing factor-1 (NRSF)/repressor element-1 silencing transcription (REST). Chromatin modification status of a number of neuronal and glial lineage restricted genes was altered upon NO-exposure. These changes coincided with gene expression alterations, demonstrating a global shift toward glial potential. Interestingly, by blocking the function of NRSF/REST, alterations in chromatin modifications were lost and the NO-induced neuronal to glial switch was suppressed. This implicates NRSF/REST as a key factor in the NPC-specific response to innate immunity and suggests a novel mechanism by which signaling from inflamed tissue promotes the formation of glial cells.


Asunto(s)
Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Animales , Diferenciación Celular/fisiología , Humanos , Células-Madre Neurales/citología , Neuronas/citología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Ratas , Transducción de Señal , Factores de Transcripción/metabolismo
8.
iScience ; 25(11): 105317, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36310582

RESUMEN

Immunotherapy for cancer that aims to promote T cell anti-tumor activity has changed current clinical practice, where some previously lethal cancers have now become treatable. However, clinical trials with low response rates have been disappointing for pancreatic ductal adenocarcinoma (PDAC). One suggested explanation is the accumulation of dominantly immunosuppressive tumor-associated macrophages and myeloid-derived suppressor cells in the tumor microenvironment (TME). Using retrospectively collected tumor specimens and transcriptomic data from PDAC, we demonstrate that expression of the scavenger receptor MARCO correlates with poor prognosis and a lymphocyte-excluding tumor phenotype. PDAC cell lines produce IL-10 and induce high expression of MARCO in myeloid cells, and this was further enhanced during hypoxic conditions. These myeloid cells suppressed effector T and natural killer (NK) cells and blocked NK cell tumor infiltration and tumor killing in a PDAC 3D-spheroid model. Anti-human MARCO (anti-hMARCO) antibody targeting triggered the repolarization of tumor-associated macrophages and activated the inflammasome machinery, resulting in IL-18 production. This in turn enhanced T cell and NK cell functions. The targeting of MARCO thus remodels the TME and represents a rational approach to make immunotherapy more efficient in PDAC patients.

9.
Nat Commun ; 11(1): 2898, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32518258

RESUMEN

The sequential generation of layer-specific cortical neurons requires radial glia cells (RGCs) to precisely balance self-renewal and lineage commitment. While specific cell-cycle phases have been associated with these decisions, the mechanisms linking the cell-cycle machinery to cell-fate commitment remain obscure. Using single-cell RNA-sequencing, we find that the strongest transcriptional signature defining multipotent RGCs is that of G2/M-phase, and particularly CYCLIN-B1/2, while lineage-committed progenitors are enriched in G1/S-phase genes, including CYCLIN-D1. These data also reveal cell-surface markers that allow us to isolate RGCs and lineage-committed progenitors, and functionally confirm the relationship between cell-cycle phase enrichment and cell fate competence. Finally, we use cortical electroporation to demonstrate that CYCLIN-B1/2 cooperate with CDK1 to maintain uncommitted RGCs by activating the NOTCH pathway, and that CYCLIN-D1 promotes differentiation. Thus, this work establishes that cell-cycle phase-specific regulators act in opposition to coordinate the self-renewal and lineage commitment of RGCs via core stem cell regulatory pathways.


Asunto(s)
Ciclina B1/fisiología , Ciclina B2/fisiología , Ciclina D1/fisiología , Regulación del Desarrollo de la Expresión Génica , Animales , Proteína Quinasa CDC2/fisiología , Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Separación Celular , Corteza Cerebral/embriología , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Neuroglía/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Células Madre/citología
10.
BMC Neurosci ; 10: 146, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20003337

RESUMEN

BACKGROUND: Parkinson's disease (PD) is caused by degeneration of dopamine (DA) neurons in the ventral midbrain (vMB) and results in severely disturbed regulation of movement. The disease inflicts considerable suffering for the affected and their families. Today, the opportunities for pharmacological treatment are meager and new technologies are needed. Previous studies have indicated that activation of the nuclear receptor Retinoid X Receptor (RXR) provides trophic support for DA neurons. Detailed investigations of these neurotrophic effects have been hampered by the lack of readily available DA neurons in vitro. The aim of this study was to further describe the potential neurotrophic actions of RXR ligands and, for this and future purposes, develop a suitable in vitro-platform using mouse embryonic stem cells (mESCs). RESULTS: We studied the potential neurotrophic effects of the RXR ligand LG100268 (LG268) and the RXR-Nurr1 ligand XCT0139508 (XCT) in neuronal cultures derived from rat primary vMB and mESCs. RXR ligands protect DA neurons from stress, such as that induced by the PD-modeling toxin 6-hydroxy dopamine (6-OHDA) and hypoxia, but not from stress induced by oxidative hydrogen peroxide (H2O2) or the excitotoxic agent kainic acid (KA). The neurotrophic effect is selective for DA neurons. DA neurons from rat primary vMB and mESCs behaved similarly, but the mESC-derived cultures contained a much higher fraction of DA cells and thus provided more accessible experimental conditions. CONCLUSIONS: RXR ligands rescue DA neurons from degeneration caused by the PD simulating 6-OHDA as well as hypoxia. Thus, RXR is a novel promising target for PD research. mESC-derived DA cells provide a valid and accessible in vitro-platform for studying PD inducing toxins and potential trophic agents.


Asunto(s)
Dopamina/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Receptores X Retinoide/fisiología , Animales , Biotransformación , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Madre Embrionarias , Ratones , Degeneración Nerviosa/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Enfermedad de Parkinson , Ratas
11.
Cancer Res ; 77(18): 4985-4997, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28687615

RESUMEN

Molecular mechanisms preventing self-renewing brain stem cells from oncogenic transformation are poorly defined. We show that the expression levels of SOX5, SOX6, and SOX21 (SOX5/6/21) transcription factors increase in stem cells of the subventricular zone (SVZ) upon oncogenic stress, whereas their expression in human glioma decreases during malignant progression. Elevated levels of SOX5/6/21 promoted SVZ cells to exit the cell cycle, whereas genetic ablation of SOX5/6/21 dramatically increased the capacity of these cells to form glioma-like tumors in an oncogene-driven mouse brain tumor model. Loss-of-function experiments revealed that SOX5/6/21 prevent detrimental hyperproliferation of oncogene expressing SVZ cells by facilitating an antiproliferative expression profile. Consistently, restoring high levels of SOX5/6/21 in human primary glioblastoma cells enabled expression of CDK inhibitors and decreased p53 protein turnover, which blocked their tumorigenic capacity through cellular senescence and apoptosis. Altogether, these results provide evidence that SOX5/6/21 play a central role in driving a tumor suppressor response in brain stem cells upon oncogenic insult. Cancer Res; 77(18); 4985-97. ©2017 AACR.


Asunto(s)
Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Glioma/patología , Células Madre Neoplásicas/patología , Factores de Transcripción SOXB2/metabolismo , Factores de Transcripción SOXD/fisiología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Senescencia Celular , Femenino , Glioma/genética , Glioma/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Oncogenes , Factores de Transcripción SOXB2/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Dev Cell ; 23(5): 1006-19, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23153497

RESUMEN

Morphogens orchestrate tissue patterning in a concentration-dependent fashion during vertebrate embryogenesis, yet little is known of how positional information provided by such signals is translated into discrete transcriptional outputs. Here we have identified and characterized cis-regulatory modules (CRMs) of genes operating downstream of graded Shh signaling and bifunctional Gli proteins in neural patterning. Unexpectedly, we find that Gli activators have a noninstructive role in long-range patterning and cooperate with SoxB1 proteins to facilitate a largely concentration-independent mode of gene activation. Instead, the opposing Gli-repressor gradient is interpreted at transcriptional levels, and, together with CRM-specific repressive input of homeodomain proteins, comprises a repressive network that translates graded Shh signaling into regional gene expression patterns. Moreover, local and long-range interpretation of Shh signaling differs with respect to CRM context sensitivity and Gli-activator dependence, and we propose that these differences provide insight into how morphogen function may have mechanistically evolved from an initially binary inductive event.


Asunto(s)
Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Neurogénesis , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Proteína con Dedos de Zinc GLI1
13.
Genes Dev ; 20(24): 3475-86, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17182872

RESUMEN

The progression of neurogenesis relies on proneural basic helix-loop-helix (bHLH) transcription factors. These factors operate in undifferentiated neural stem cells and induce cell cycle exit and the initiation of a neurogenic program. However, the transient expression of proneural bHLH proteins in neural progenitors indicates that expression of neuronal traits must rely on previously unexplored mechanisms operating downstream from proneural bHLH proteins. Here we show that the HMG-box transcription factors Sox4 and Sox11 are of critical importance, downstream from proneural bHLH proteins, for the establishment of pan-neuronal protein expression. Examination of a neuronal gene promoter reveals that Sox4 and Sox11 exert their functions as transcriptional activators. Interestingly, the capacity of Sox4 and Sox11 to induce the expression of neuronal traits is independent of mechanisms regulating the exit of neural progenitors from the cell cycle. The transcriptional repressor protein REST/NRSF has been demonstrated to block neuronal gene expression in undifferentiated neural cells. We now show that REST/NRSF restricts the expression of Sox4 and Sox11, explaining how REST/NRSF can prevent precocious expression of neuronal proteins. Together, these findings demonstrate a central regulatory role of Sox4 and Sox11 during neuronal maturation and mechanistically separate cell cycle withdrawal from the establishment of neuronal properties.


Asunto(s)
Proteínas Aviares/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/fisiología , Neuronas/citología , Activación Transcripcional , Animales , Proteínas Aviares/genética , Diferenciación Celular , Embrión de Pollo , Proteínas HMGB/genética , Ratones , Neuronas/metabolismo , Regiones Promotoras Genéticas
14.
J Neurochem ; 97(5): 1403-11, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16638018

RESUMEN

The orphan nuclear receptor Nurr1 is expressed in the developing and adult central nervous system. Previous studies have shown that Nurr1 is essential for the generation of midbrain dopamine neurons. Furthermore, Nurr1 is critical for respiratory functions associated with the brain stem. Very few Nurr1 regulated genes have been identified and it remains unclear how Nurr1 influences the function and development of neurons. To identify novel Nurr1 target genes we have searched for regulated genes in the dopaminergic MN9D cell line. These experiments identified Neuropilin-1 (Nrp1), a receptor protein involved in axon guidance and angiogenesis, as a novel Nurr1 target gene. Nrp1 expression was rapidly up-regulated by Nurr1 in MN9D cells and in situ hybridization analysis showed that Nrp1 was coexpressed with Nurr1 in the brain stem dorsal motor nucleus. Importantly, Nrp1 expression was down-regulated in this area in Nurr1 null mice. Moreover, two functional Nurr1 binding sites were identified in the Nrp1 promoter and Nurr1 was found to be recruited to these sites in MN9D cells, further supporting that Nrp1 is a direct downstream target of Nurr1. Taken together, our findings suggest that Nurr1 might influence the processes of axon guidance and/or angiogenesis via the regulation of Nrp1 expression.


Asunto(s)
Tronco Encefálico/embriología , Tronco Encefálico/metabolismo , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Neuropilina-1/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión/genética , Tronco Encefálico/citología , Línea Celular , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/fisiología , Femenino , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Ratones , Ratones Noqueados , Neovascularización Fisiológica/fisiología , Neuropilina-1/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética , Regulación hacia Arriba/fisiología , Nervio Vago/citología , Nervio Vago/embriología , Nervio Vago/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA