Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(2)2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36674677

RESUMEN

DNA sequence variants (single nucleotide polymorphisms or variants, SNPs/SNVs; copy number variants, CNVs) associated to neurodevelopmental disorders (NDD) and traits often map on putative transcriptional regulatory elements, including, in particular, enhancers. However, the genes controlled by these enhancers remain poorly defined. Traditionally, the activity of a given enhancer, and the effect of its possible alteration associated to the sequence variants, has been thought to influence the nearest gene promoter. However, the obtainment of genome-wide long-range interaction maps in neural cells chromatin challenged this view, showing that a given enhancer is very frequently not connected to the nearest promoter, but to a more distant one, skipping genes in between. In this Perspective, we review some recent papers, who generated long-range interaction maps (by HiC, RNApolII ChIA-PET, Capture-HiC, or PLACseq), and overlapped the identified long-range interacting DNA segments with DNA sequence variants associated to NDD (such as schizophrenia, bipolar disorder and autism) and traits (intelligence). This strategy allowed to attribute the function of enhancers, hosting the NDD-related sequence variants, to a connected gene promoter lying far away on the linear chromosome map. Some of these enhancer-connected genes had indeed been already identified as contributive to the diseases, by the identification of mutations within the gene's protein-coding regions (exons), validating the approach. Significantly, however, the connected genes also include many genes that were not previously found mutated in their exons, pointing to novel candidate contributors to NDD and traits. Thus, long-range interaction maps, in combination with DNA variants detected in association with NDD, can be used as "pointers" to identify novel candidate disease-relevant genes. Functional manipulation of the long-range interaction network involving enhancers and promoters by CRISPR-Cas9-based approaches is beginning to probe for the functional significance of the identified interactions, and the enhancers and the genes involved, improving our understanding of neural development and its pathology.


Asunto(s)
Cromatina , Trastornos del Neurodesarrollo , Humanos , Cromatina/genética , Elementos de Facilitación Genéticos , ADN , Regiones Promotoras Genéticas , Trastornos del Neurodesarrollo/genética , Estudio de Asociación del Genoma Completo
2.
Stem Cells ; 39(8): 1107-1119, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33739574

RESUMEN

The Sox2 transcription factor is necessary for the long-term self-renewal of neural stem cells (NSCs). Its mechanism of action is still poorly defined. To identify molecules regulated by Sox2, and acting in mouse NSC maintenance, we transduced, into Sox2-deleted NSC, genes whose expression is strongly downregulated following Sox2 loss (Fos, Jun, Egr2), individually or in combination. Fos alone rescued long-term proliferation, as shown by in vitro cell growth and clonal analysis. Furthermore, pharmacological inhibition by T-5224 of FOS/JUN AP1 complex binding to its targets decreased cell proliferation and expression of the putative target Suppressor of cytokine signaling 3 (Socs3). Additionally, Fos requirement for efficient long-term proliferation was demonstrated by the reduction of NSC clones capable of long-term expansion following CRISPR/Cas9-mediated Fos inactivation. Previous work showed that the Socs3 gene is strongly downregulated following Sox2 deletion, and its re-expression by lentiviral transduction rescues long-term NSC proliferation. Fos appears to be an upstream regulator of Socs3, possibly together with Jun and Egr2; indeed, Sox2 re-expression in Sox2-deleted NSC progressively activates both Fos and Socs3 expression; in turn, Fos transduction activates Socs3 expression. Based on available SOX2 ChIPseq and ChIA-PET data, we propose a model whereby Sox2 is a direct activator of both Socs3 and Fos, as well as possibly Jun and Egr2; furthermore, we provide direct evidence for FOS and JUN binding on Socs3 promoter, suggesting direct transcriptional regulation. These results provide the basis for developing a model of a network of interactions, regulating critical effectors of NSC proliferation and long-term maintenance.


Asunto(s)
Células-Madre Neurales , Proteínas Proto-Oncogénicas c-fos , Factores de Transcripción SOXB1 , Animales , Proliferación Celular/genética , Autorrenovación de las Células/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Ratones , Células-Madre Neurales/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
3.
Int J Mol Sci ; 23(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35887306

RESUMEN

Non-coding variation in complex human disease has been well established by genome-wide association studies, and is thought to involve regulatory elements, such as enhancers, whose variation affects the expression of the gene responsible for the disease. The regulatory elements often lie far from the gene they regulate, or within introns of genes differing from the regulated gene, making it difficult to identify the gene whose function is affected by a given enhancer variation. Enhancers are connected to their target gene promoters via long-range physical interactions (loops). In our study, we re-mapped, onto the human genome, more than 10,000 enhancers connected to promoters via long-range interactions, that we had previously identified in mouse brain-derived neural stem cells by RNApolII-ChIA-PET analysis, coupled to ChIP-seq mapping of DNA/chromatin regions carrying epigenetic enhancer marks. These interactions are thought to be functionally relevant. We discovered, in the human genome, thousands of DNA regions syntenic with the interacting mouse DNA regions (enhancers and connected promoters). We further annotated these human regions regarding their overlap with sequence variants (single nucleotide polymorphisms, SNPs; copy number variants, CNVs), that were previously associated with neurodevelopmental disease in humans. We document various cases in which the genetic variant, associated in humans to neurodevelopmental disease, affects an enhancer involved in long-range interactions: SNPs, previously identified by genome-wide association studies to be associated with schizophrenia, bipolar disorder, and intelligence, are located within our human syntenic enhancers, and alter transcription factor recognition sites. Similarly, CNVs associated to autism spectrum disease and other neurodevelopmental disorders overlap with our human syntenic enhancers. Some of these enhancers are connected (in mice) to homologs of genes already associated to the human disease, strengthening the hypothesis that the gene is indeed involved in the disease. Other enhancers are connected to genes not previously associated with the disease, pointing to their possible pathogenetic involvement. Our observations provide a resource for further exploration of neural disease, in parallel with the now widespread genome-wide identification of DNA variants in patients with neural disease.


Asunto(s)
Células-Madre Neurales , Trastornos del Neurodesarrollo , Animales , Cromatina/genética , Elementos de Facilitación Genéticos/genética , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Trastornos del Neurodesarrollo/genética , Regiones Promotoras Genéticas/genética
4.
Glia ; 69(3): 579-593, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32975900

RESUMEN

Cancer stem cells (CSC) are essential for tumorigenesis. The transcription factor Sox2 is overexpressed in brain gliomas, and is essential to maintain CSC. In mouse high-grade glioma pHGG cells in culture, Sox2 deletion causes cell proliferation arrest and inability to reform tumors after transplantation in vivo; in Sox2-deleted cells, 134 genes are derepressed. To identify genes mediating Sox2 deletion effects, we overexpressed into pHGG cells nine among the most derepressed genes, and identified four genes, Ebf1, Hey2, Zfp423, and Cdkn2b, that strongly reduced cell proliferation in vitro and brain tumorigenesis in vivo. CRISPR/Cas9 mutagenesis of each gene, individually or in combination (Ebf1 + Cdkn2b), significantly antagonized the proliferation arrest caused by Sox2 deletion. The same genes also repressed clonogenicity in primary human glioblastoma-derived CSC-like lines. These experiments identify a network of critical tumor suppressive Sox2-targets whose inhibition by Sox2 is involved in glioma CSC maintenance, defining new potential therapeutic targets.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Oligodendroglioma , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Neoplasias Encefálicas/genética , Carcinogénesis/genética , Línea Celular Tumoral , Regulación hacia Abajo , Glioma/genética , Ratones , Células Madre Neoplásicas/metabolismo , Proteínas Represoras , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transactivadores
5.
Development ; 145(2)2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29352015

RESUMEN

The transcription factor Sox2 is necessary to maintain pluripotency of embryonic stem cells, and to regulate neural development. Neurogenesis in the vertebrate olfactory epithelium persists from embryonic stages through adulthood. The role Sox2 plays for the development of the olfactory epithelium and neurogenesis within has, however, not been determined. Here, by analysing Sox2 conditional knockout mouse embryos and chick embryos deprived of Sox2 in the olfactory epithelium using CRISPR-Cas9, we show that Sox2 activity is crucial for the induction of the neural progenitor gene Hes5 and for subsequent differentiation of the neuronal lineage. Our results also suggest that Sox2 activity promotes the neurogenic domain in the nasal epithelium by restricting Bmp4 expression. The Sox2-deficient olfactory epithelium displays diminished cell cycle progression and proliferation, a dramatic increase in apoptosis and finally olfactory pit atrophy. Moreover, chromatin immunoprecipitation data show that Sox2 directly binds to the Hes5 promoter in both the PNS and CNS. Taken together, our results indicate that Sox2 is essential to establish, maintain and expand the neuronal progenitor pool by suppressing Bmp4 and upregulating Hes5 expression.


Asunto(s)
Proteínas Aviares/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas Morfogenéticas Óseas/metabolismo , Neurogénesis/fisiología , Mucosa Olfatoria/embriología , Mucosa Olfatoria/metabolismo , Proteínas Represoras/genética , Factores de Transcripción SOXB1/metabolismo , Animales , Apoptosis , Proteínas Aviares/deficiencia , Proteínas Aviares/genética , Secuencia de Bases , Sitios de Unión/genética , Proteína Morfogenética Ósea 4/metabolismo , Ciclo Celular , Linaje de la Célula , Proliferación Celular , Embrión de Pollo , Femenino , Técnicas de Inactivación de Genes , Ratones , Ratones Noqueados , Neurogénesis/genética , Embarazo , Regiones Promotoras Genéticas , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Regulación hacia Arriba
6.
Nature ; 504(7479): 306-310, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24213634

RESUMEN

In multicellular organisms, transcription regulation is one of the central mechanisms modelling lineage differentiation and cell-fate determination. Transcription requires dynamic chromatin configurations between promoters and their corresponding distal regulatory elements. It is believed that their communication occurs within large discrete foci of aggregated RNA polymerases termed transcription factories in three-dimensional nuclear space. However, the dynamic nature of chromatin connectivity has not been characterized at the genome-wide level. Here, through a chromatin interaction analysis with paired-end tagging approach using an antibody that primarily recognizes the pre-initiation complexes of RNA polymerase II, we explore the transcriptional interactomes of three mouse cells of progressive lineage commitment, including pluripotent embryonic stem cells, neural stem cells and neurosphere stem/progenitor cells. Our global chromatin connectivity maps reveal approximately 40,000 long-range interactions, suggest precise enhancer-promoter associations and delineate cell-type-specific chromatin structures. Analysis of the complex regulatory repertoire shows that there are extensive colocalizations among promoters and distal-acting enhancers. Most of the enhancers associate with promoters located beyond their nearest active genes, indicating that the linear juxtaposition is not the only guiding principle driving enhancer target selection. Although promoter-enhancer interactions exhibit high cell-type specificity, promoters involved in interactions are found to be generally common and mostly active among different cells. Chromatin connectivity networks reveal that the pivotal genes of reprogramming functions are transcribed within physical proximity to each other in embryonic stem cells, linking chromatin architecture to coordinated gene expression. Our study sets the stage for the full-scale dissection of spatial and temporal genome structures and their roles in orchestrating development.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Animales , Línea Celular , Linaje de la Célula , Células Madre Embrionarias/metabolismo , Hibridación Fluorescente in Situ , Ratones , Células-Madre Neurales/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética/genética , Pez Cebra/genética
7.
Int J Mol Sci ; 20(18)2019 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-31540269

RESUMEN

The Sox2 transcription factor, encoded by a gene conserved in animal evolution, has become widely known because of its functional relevance for stem cells. In the developing nervous system, Sox2 is active in neural stem cells, and important for their self-renewal; differentiation to neurons and glia normally involves Sox2 downregulation. Recent evidence, however, identified specific types of fully differentiated neurons and glia that retain high Sox2 expression, and critically require Sox2 function, as revealed by functional studies in mouse and in other animals. Sox2 was found to control fundamental aspects of the biology of these cells, such as the development of correct neuronal connectivity. Sox2 downstream target genes identified within these cell types provide molecular mechanisms for cell-type-specific Sox2 neuronal and glial functions. SOX2 mutations in humans lead to a spectrum of nervous system defects, involving vision, movement control, and cognition; the identification of neurons and glia requiring Sox2 function, and the investigation of Sox2 roles and molecular targets within them, represents a novel perspective for the understanding of the pathogenesis of these defects.


Asunto(s)
Células-Madre Neurales/citología , Neuroglía/citología , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Animales , Diferenciación Celular , Autorrenovación de las Células , Regulación hacia Abajo , Humanos , Ratones , Mutación , Células-Madre Neurales/metabolismo , Neurogénesis , Neuroglía/metabolismo , Transducción de Señal
8.
Glia ; 66(9): 1929-1946, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29732603

RESUMEN

Sox2 is a transcription factor active in the nervous system, within different cell types, ranging from radial glia neural stem cells to a few specific types of differentiated glia and neurons. Mutations in the human SOX2 transcription factor gene cause various central nervous system (CNS) abnormalities, involving hippocampus and eye defects, as well as ataxia. Conditional Sox2 mutation in mouse, with different Cre transgenes, previously recapitulated different essential features of the disease, such as hippocampus and eye defects. In the cerebellum, Sox2 is active from early embryogenesis in the neural progenitors of the cerebellar primordium; Sox2 expression is maintained, postnatally, within Bergmann glia (BG), a differentiated cell type essential for Purkinje neurons functionality and correct motor control. By performing Sox2 Cre-mediated ablation in the developing and postnatal mouse cerebellum, we reproduced ataxia features. Embryonic Sox2 deletion (with Wnt1Cre) leads to reduction of the cerebellar vermis, known to be commonly related to ataxia, preceded by deregulation of Otx2 and Gbx2, critical regulators of vermis development. Postnatally, BG is progressively disorganized, mislocalized, and reduced in mutants. Sox2 postnatal deletion, specifically induced in glia (with GLAST-CreERT2), reproduces the BG defect, and causes (milder) ataxic features. Our results define a role for Sox2 in cerebellar function and development, and identify a functional requirement for Sox2 within postnatal BG, of potential relevance for ataxia in mouse mutants, and in human patients.


Asunto(s)
Ataxia/metabolismo , Vermis Cerebeloso/crecimiento & desarrollo , Vermis Cerebeloso/metabolismo , Neuroglía/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Animales Recién Nacidos , Ataxia/patología , Células Cultivadas , Vermis Cerebeloso/patología , Regulación de la Expresión Génica/fisiología , Ácido Glutámico/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones Transgénicos , Mutación , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuroglía/patología , Factores de Transcripción Otx/metabolismo , Factores de Transcripción SOXB1/genética , Transmisión Sináptica/fisiología
9.
EMBO J ; 32(14): 1990-2000, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23673358

RESUMEN

While the Polycomb complex is known to regulate cell identity in ES cells, its role in controlling tissue-specific stem cells is not well understood. Here we show that removal of Ezh1 and Ezh2, key Polycomb subunits, from mouse skin results in a marked change in fate determination in epidermal progenitor cells, leading to an increase in the number of lineage-committed Merkel cells, a specialized subtype of skin cells involved in mechanotransduction. By dissecting the genetic mechanism, we showed that the Polycomb complex restricts differentiation of epidermal progenitor cells by repressing the transcription factor Sox2. Ablation of Sox2 results in a dramatic loss of Merkel cells, indicating that Sox2 is a critical regulator of Merkel cell specification. We show that Sox2 directly activates Atoh1, the obligate regulator of Merkel cell differentiation. Concordantly, ablation of Sox2 attenuated the Ezh1/2-null phenotype, confirming the importance of Polycomb-mediated repression of Sox2 in maintaining the epidermal progenitor cell state. Together, these findings define a novel regulatory network by which the Polycomb complex maintains the progenitor cell state and governs differentiation in vivo.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Células de Merkel/citología , Células de Merkel/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Complejo Represivo Polycomb 2/deficiencia , Complejo Represivo Polycomb 2/genética , Embarazo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
EMBO J ; 32(16): 2231-47, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23892456

RESUMEN

Embryonic stem (ES) cell self-renewal efficiency is determined by the Nanog protein level. However, the protein partners of Nanog that function to direct self-renewal are unclear. Here, we identify a Nanog interactome of over 130 proteins including transcription factors, chromatin modifying complexes, phosphorylation and ubiquitination enzymes, basal transcriptional machinery members, and RNA processing factors. Sox2 was identified as a robust interacting partner of Nanog. The purified Nanog-Sox2 complex identified a DNA recognition sequence present in multiple overlapping Nanog/Sox2 ChIP-Seq data sets. The Nanog tryptophan repeat region is necessary and sufficient for interaction with Sox2, with tryptophan residues required. In Sox2, tyrosine to alanine mutations within a triple-repeat motif (S X T/S Y) abrogates the Nanog-Sox2 interaction, alters expression of genes associated with the Nanog-Sox2 cognate sequence, and reduces the ability of Sox2 to rescue ES cell differentiation induced by endogenous Sox2 deletion. Substitution of the tyrosines with phenylalanine rescues both the Sox2-Nanog interaction and efficient self-renewal. These results suggest that aromatic stacking of Nanog tryptophans and Sox2 tyrosines mediates an interaction central to ES cell self-renewal.


Asunto(s)
Proliferación Celular , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Factores de Transcripción SOXB1/metabolismo , Animales , Ensayo de Unidades Formadoras de Colonias , Células Madre Embrionarias/metabolismo , Immunoblotting , Inmunoprecipitación , Ratones , Proteína Homeótica Nanog , Plásmidos/genética , Mapeo de Interacción de Proteínas , Técnica SELEX de Producción de Aptámeros , Triptófano/metabolismo , Tirosina/metabolismo
11.
Development ; 140(6): 1250-61, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23444355

RESUMEN

The Sox2 transcription factor is active in stem/progenitor cells throughout the developing vertebrate central nervous system. However, its conditional deletion at E12.5 in mouse causes few brain developmental problems, with the exception of the postnatal loss of the hippocampal radial glia stem cells and the dentate gyrus. We deleted Sox2 at E9.5 in the telencephalon, using a Bf1-Cre transgene. We observed embryonic brain defects that were particularly severe in the ventral, as opposed to the dorsal, telencephalon. Important tissue loss, including the medial ganglionic eminence (MGE), was detected at E12.5, causing the subsequent impairment of MGE-derived neurons. The defect was preceded by loss of expression of the essential ventral determinants Nkx2.1 and Shh, and accompanied by ventral spread of dorsal markers. This phenotype is reminiscent of that of mice mutant for the transcription factor Nkx2.1 or for the Shh receptor Smo. Nkx2.1 is known to mediate the initial activation of ventral telencephalic Shh expression. A partial rescue of the normal phenotype at E14.5 was obtained by administration of a Shh agonist. Experiments in Medaka fish indicate that expression of Nkx2.1 is regulated by Sox2 in this species also. We propose that Sox2 contributes to Nkx2.1 expression in early mouse development, thus participating in the region-specific activation of Shh, thereby mediating ventral telencephalic patterning induction.


Asunto(s)
Tipificación del Cuerpo/genética , Desarrollo Embrionario/genética , Proteínas Hedgehog/genética , Proteínas Nucleares/genética , Factores de Transcripción SOXB1/fisiología , Telencéfalo/embriología , Factores de Transcripción/genética , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/metabolismo , Embarazo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Telencéfalo/metabolismo , Factor Nuclear Tiroideo 1 , Factores de Transcripción/metabolismo , Activación Transcripcional/genética
12.
Development ; 139(2): 397-410, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22186729

RESUMEN

The cellular origin and molecular mechanisms regulating pigmentation of head and neck are largely unknown. Melanocyte specification is controlled by the transcriptional activity of Mitf, but no general logic has emerged to explain how Mitf and progenitor transcriptional activities consolidate melanocyte and progenitor cell fates. We show that cranial melanocytes arise from at least two different cellular sources: initially from nerve-associated Schwann cell precursors (SCPs) and later from a cellular source that is independent of nerves. Unlike the midbrain-hindbrain cluster from which melanoblasts arise independently of nerves, a large center of melanocytes in and around cranial nerves IX-X is derived from SCPs, as shown by genetic cell-lineage tracing and analysis of ErbB3-null mutant mice. Conditional gain- and loss-of-function experiments show genetically that cell fates in the neural crest involve both the SRY transcription factor Sox2 and Mitf, which consolidate an SCP progenitor or melanocyte fate by cross-regulatory interactions. A gradual downregulation of Sox2 in progenitors during development permits the differentiation of both neural crest- and SCP-derived progenitors into melanocytes, and an initial small pool of nerve-associated melanoblasts expands in number and disperses under the control of endothelin receptor B (Ednrb) and Wnt5a signaling.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Melanocitos/citología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Cresta Neural/embriología , Pigmentación/fisiología , Factores de Transcripción SOXB1/metabolismo , Animales , Inmunoprecipitación de Cromatina , Embrión de Mamíferos/embriología , Imagenología Tridimensional , Inmunohistoquímica , Hibridación in Situ , Melanocitos/metabolismo , Ratones , Cresta Neural/metabolismo , Plásmidos/genética , ARN Interferente Pequeño/genética , Receptores de Endotelina/metabolismo , Células de Schwann/citología , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , Proteína Wnt-5a
13.
Development ; 138(5): 971-81, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21303850

RESUMEN

The mammalian foregut gives rise to the dorsally located esophagus and stomach and the ventrally located trachea and lung. Proper patterning and morphogenesis of the common foregut tube and its derived organs is essential for viability of the organism at birth. Here, we show that conditional inactivation of BMP type I receptor genes Bmpr1a and Bmpr1b (Bmpr1a;b) in the ventral endoderm leads to tracheal agenesis and ectopic primary bronchi. Molecular analyses of these mutants reveal a reduction of ventral endoderm marker NKX2-1 and an expansion of dorsal markers SOX2 and P63 into the prospective trachea and primary bronchi. Subsequent genetic experiments show that activation of canonical WNT signaling, previously shown to induce ectopic respiratory fate in otherwise wild-type mice, is incapable of promoting respiratory fate in the absence of Bmpr1a;b. Furthermore, we find that inactivation of Sox2 in Bmpr1a;b mutants does not suppress ectopic lung budding but does rescue trachea formation and NKX2-1 expression. Together, our data suggest that signaling through BMPR1A;B performs at least two roles in early respiratory development: first, it promotes tracheal formation through repression of Sox2; and second, it restricts the site of lung bud initiation.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Sistema Digestivo/embriología , Sistema Respiratorio/embriología , Factores de Transcripción SOXB1/antagonistas & inhibidores , Tráquea/embriología , Animales , Bronquios/anomalías , Bronquios/embriología , Constricción Patológica/etiología , Ratones , Tráquea/anomalías
14.
Stem Cells ; 31(7): 1408-21, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23553930

RESUMEN

Sox2 is a pluripotency-conferring gene expressed in primordial germ cells (PGCs) and postnatal oocytes, but the role it plays during germ cell development and early embryogenesis is unknown. Since Sox2 ablation causes early embryonic lethality shortly after blastocyst implantation, we generated mice bearing Sox2-conditional deletion in germ cells at different stages of their development through the Cre/loxP recombination system. Embryos lacking Sox2 in PGCs show a dramatic decrease of germ cell numbers at the time of their specification. At later stages, we found that Sox2 is strictly required for PGC proliferation. On the contrary, Sox2 deletion in meiotic oocytes does not impair postnatal oogenesis and early embryogenesis, indicating that it is not essential for oocyte maturation or for zygotic development. We also show that Sox2 regulates Kit expression in PGCs and binds to discrete transcriptional regulatory sequences of this gene, which is known to be important for PGCs survival and proliferation. Sox2 also stimulates the expression of Zfp148, which is required for normal development of fetal germ cells, and Rif1, a potential regulator of PGC pluripotency.


Asunto(s)
Células Germinativas/citología , Factores de Transcripción SOXB1/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Expresión Génica , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Oocitos/citología , Oocitos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Stem Cells ; 30(5): 817-32, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22311806

RESUMEN

The invasive nature of glioblastoma (GBM) is one important reason for treatment failure. GBM stem/progenitor cells retain the migratory ability of normal neural stem/progenitor cells and infiltrate the brain parenchyma. Here, we identify Rai (ShcC/N-Shc), a member of the family of Shc-like adaptor proteins, as a new regulator of migration of normal and cancer stem/progenitor cells. Rai is expressed in neurogenic areas of the brain and its knockdown impairs progenitor migration to the olfactory bulb. Its expression is retained in GBM stem/progenitor cells where it exerts the same promigratory activity. Rai silencing in cancer stem/progenitor cells isolated from different patients causes significant decrease in cell migration and invasion, both in vitro and in vivo, providing survival benefit. Rai depletion is associated with alteration of multiple-signaling pathways, yet it always leads to reduced expression of proinvasive genes.


Asunto(s)
Movimiento Celular , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Animales , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Ratones Noqueados , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/patología , Bulbo Olfatorio , Proteínas Adaptadoras de la Señalización Shc/genética , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src , Células Tumorales Cultivadas
16.
Mol Cell Endocrinol ; 563: 111864, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36690169

RESUMEN

Prenatal exposure to synthetic glucocorticoids (sGCs) reprograms brain development and predisposes the developing fetus towards potential adverse neurodevelopmental outcomes. Using a mouse model of sGC administration, previous studies show that these changes are accompanied by sexually dimorphic alterations in the transcriptome of neural stem and progenitor cells (NSPCs) derived from the embryonic telencephalon. Because cell type-specific gene expression profiles tightly regulate cell fate decisions and are controlled by a flexible landscape of chromatin domains upon which transcription factors and enhancer elements act, we multiplexed data from four genome-wide assays: RNA-seq, ATAC-seq (assay for transposase accessible chromatin followed by genome wide sequencing), dual cross-linking ChIP-seq (chromatin immunoprecipitation followed by genome wide sequencing), and microarray gene expression to identify novel relationships between gene regulation, chromatin structure, and genomic glucocorticoid receptor (GR) action in NSPCs. These data reveal that GR binds preferentially to predetermined regions of accessible chromatin to influence gene programming and cell fate decisions. In addition, we identify SOX2 as a transcription factor that impacts the genomic response of select GR target genes to sGCs (i.e., dexamethasone) in NSPCs.


Asunto(s)
Glucocorticoides , Células-Madre Neurales , Femenino , Embarazo , Cromatina/metabolismo , Regulación de la Expresión Génica , Genómica , Glucocorticoides/farmacología , Glucocorticoides/metabolismo , Células-Madre Neurales/metabolismo , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Ratones , Células Madre Embrionarias de Ratones
17.
Cells ; 11(10)2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35626641

RESUMEN

SOX2 is a transcription factor conserved throughout vertebrate evolution, whose expression marks the central nervous system from the earliest developmental stages. In humans, SOX2 mutation leads to a spectrum of CNS defects, including vision and hippocampus impairments, intellectual disability, and motor control problems. Here, we review how conditional Sox2 knockout (cKO) in mouse with different Cre recombinases leads to very diverse phenotypes in different regions of the developing and postnatal brain. Surprisingly, despite the widespread expression of Sox2 in neural stem/progenitor cells of the developing neural tube, some regions (hippocampus, ventral forebrain) appear much more vulnerable than others to Sox2 deletion. Furthermore, the stage of Sox2 deletion is also a critical determinant of the resulting defects, pointing to a stage-specificity of SOX2 function. Finally, cKOs illuminate the importance of SOX2 function in different cell types according to the different affected brain regions (neural precursors, GABAergic interneurons, glutamatergic projection neurons, Bergmann glia). We also review human genetics data regarding the brain defects identified in patients carrying mutations within human SOX2 and examine the parallels with mouse mutants. Functional genomics approaches have started to identify SOX2 molecular targets, and their relevance for SOX2 function in brain development and disease will be discussed.


Asunto(s)
Células-Madre Neurales , Neuroglía , Factores de Transcripción SOXB1/metabolismo , Animales , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Humanos , Ratones , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción/metabolismo
18.
Cell Rep ; 38(5): 110313, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108528

RESUMEN

The adult neurogenic niche in the hippocampus is maintained through activation of reversibly quiescent neural stem cells (NSCs) with radial glia-like morphology (RGLs). Here, we show that the expression of SoxD transcription factors Sox5 and Sox6 is enriched in activated RGLs. Using inducible deletion of Sox5 or Sox6 in the adult mouse brain, we show that both genes are required for RGL activation and the generation of new neurons. Conversely, Sox5 overexpression in cultured NSCs interferes with entry in quiescence. Mechanistically, expression of the proneural protein Ascl1 (a key RGL regulator) is severely downregulated in SoxD-deficient RGLs, and Ascl1 transcription relies on conserved Sox motifs. Additionally, loss of Sox5 hinders the RGL activation driven by neurogenic stimuli such as environmental enrichment. Altogether, our data suggest that SoxD genes are key mediators in the transition of adult RGLs from quiescence to an activated mitotic state under physiological situations.


Asunto(s)
Células Madre Adultas/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción SOXD/metabolismo , Animales , Diferenciación Celular/fisiología , Hipocampo/metabolismo , Ratones Transgénicos , Neurogénesis/fisiología , Factores de Transcripción SOXD/genética , Factores de Transcripción/metabolismo
19.
J Neurosci ; 30(44): 14635-48, 2010 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21048121

RESUMEN

Sox2 is expressed by neural stem and progenitor cells, and a sox2 enhancer identifies these cells in the forebrains of both fetal and adult transgenic mouse reporters. We found that an adenovirus encoding EGFP placed under the regulatory control of a 0.4 kb sox2 core enhancer selectively identified multipotential and self-renewing neural progenitor cells in dissociates of human fetal forebrain. Upon EGFP-based fluorescence-activated cell sorting (FACS), the E/sox2:EGFP(+) isolates were propagable for up to 1 year in vitro, and remained multilineage competent throughout. E/sox2:EGFP(+) cells expressed more telomerase enzymatic activity than matched E/sox2:EGFP-depleted populations, and maintained their telomeric lengths with successive passage. Gene expression analysis of E/sox2:EGFP-sorted neural progenitor cells, normalized to the unsorted forebrain dissociates from which they derived, revealed marked overexpression of genes within the notch and wnt pathways, and identified multiple elements of each pathway that appear selective to human neural progenitors. Sox2 enhancer-based FACS thus permits the prospective identification and direct isolation of a telomerase-active population of neural stem cells from the human fetal forebrain, and the elucidation of both the transcriptome and dominant signaling pathways of these critically important cells.


Asunto(s)
Células Madre Embrionarias/citología , Elementos de Facilitación Genéticos/genética , Citometría de Flujo/métodos , Células-Madre Neurales/citología , Factores de Transcripción SOXB1/genética , Telomerasa/biosíntesis , Linaje de la Célula/genética , Separación Celular/métodos , Células Cultivadas , Células Madre Embrionarias/clasificación , Células Madre Embrionarias/enzimología , Feto , Humanos , Células-Madre Neurales/clasificación , Células-Madre Neurales/enzimología , Estudios Prospectivos , Telomerasa/genética
20.
Glia ; 59(11): 1588-99, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21766338

RESUMEN

Radial-glia-like neural stem (NS) cells may be derived from neural tissues or via differentiation of pluripotent embryonic stem (ES) cells. However, the mechanisms controlling NS cell propagation and differentiation are not yet fully understood. Here we investigated the roles of Sox2 and Pax6, transcription factors widely expressed in central nervous system (CNS) progenitors, in mouse NS cells. Conditional deletion of either Sox2 or Pax6 in forebrain-derived NS cells reduced their clonogenicity in a gene dosage-dependent manner. Cells heterozygous for either gene displayed moderate proliferative defects, which may relate to human pathologies attributed to SOX2 or PAX6 deficiencies. In the complete absence of Sox2, cells exited the cell cycle with concomitant downregulation of neural progenitor markers Nestin and Blbp. This occurred despite expression of the close relative Sox3. Ablation of Pax6 also caused major proliferative defects. However, a subpopulation of cells was able to expand continuously without Pax6. These Pax6-null cells retained progenitor markers but had altered morphology. They exhibited compromised differentiation into astrocytes and oligodendrocytes, highlighting that the role of Pax6 extends beyond neurogenic competence. Overall these findings indicate that Sox2 and Pax6 are both critical for self-renewal of differentiation-competent radial glia-like NS cells.


Asunto(s)
Proliferación Celular , Proteínas del Ojo/fisiología , Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Neuroglía/fisiología , Factores de Transcripción Paired Box/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Astrocitos/fisiología , Astrocitos/ultraestructura , Western Blotting , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Proteínas del Ojo/genética , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos/fisiología , Citometría de Flujo , Dosificación de Gen , Proteínas de Homeodominio/genética , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/fisiología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/fisiología , Nestina , Células-Madre Neurales/ultraestructura , Neuroglía/ultraestructura , Oligodendroglía/fisiología , Oligodendroglía/ultraestructura , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Factores de Transcripción SOXB1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA