Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32487737

RESUMEN

The intrinsic mechanisms that link extracellular signalling to the onset of neural differentiation are not well understood. In pluripotent mouse cells, BMP blocks entry into the neural lineage via transcriptional upregulation of inhibitor of differentiation (Id) factors. We have previously identified the major binding partner of Id proteins in pluripotent cells as the basic helix-loop-helix (bHLH) transcription factor (TF) E2A. Id1 can prevent E2A from forming heterodimers with bHLH TFs or from forming homodimers. Here, we show that overexpression of a forced E2A homodimer is sufficient to drive robust neural commitment in pluripotent cells, even under non-permissive conditions. Conversely, we find that E2A null cells display a defect in their neural differentiation capacity. E2A acts as an upstream activator of neural lineage genes, including Sox1 and Foxd4, and as a repressor of Nodal signalling. Our results suggest a crucial role for E2A in establishing neural lineage commitment in pluripotent cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Neuronas/metabolismo , Regiones no Traducidas 3' , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sistemas CRISPR-Cas/genética , Linaje de la Célula , Autorrenovación de las Células , Dimerización , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Neuronas/citología , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transcriptoma , Regulación hacia Arriba
2.
Development ; 147(11)2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32439758

RESUMEN

We previously identified the cyclin dependent kinase Cdk8 as a putative silencing factor for Xist To investigate its role in X inactivation, we engineered a Cdk8 mutation in mouse embryonic stem cells (ESCs) carrying an inducible system for studying Xist function. We found that Xist repressed X-linked genes at half of the expression level in Cdk8 mutant cells, whereas they were almost completely silenced in the controls. Lack of Cdk8 impaired Ezh2 recruitment and the establishment of histone H3 lysine 27 tri-methylation but not PRC1 recruitment by Xist Transgenic expression of wild-type but not catalytically inactive Cdk8 restored efficient gene repression and PRC2 recruitment. Mutation of the paralogous kinase Cdk19 did not affect Xist function, and combined mutations of Cdk8 and Cdk19 resembled the Cdk8 mutation. In mice, a Cdk8 mutation caused post-implantation lethality. We observed that homozygous Cdk8 mutant female embryos showed a greater developmental delay than males on day 10.5. Together with the inefficient repression of X-linked genes in differentiating Cdk8 mutant female ESCs, these data show a requirement for Cdk8 in the initiation of X inactivation.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/metabolismo , Histonas/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Quinasa 8 Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Embrión de Mamíferos , Desarrollo Embrionario , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Edición Génica , Regulación del Desarrollo de la Expresión Génica , Masculino , Metilación , Ratones , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Mutagénesis , Complejo Represivo Polycomb 2/metabolismo , ARN Guía de Kinetoplastida/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética
3.
Sci Rep ; 10(1): 6751, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32317665

RESUMEN

SOX3 is a transcription factor expressed within the developing and adult nervous system where it mostly functions to help maintain neural precursors. Sox3 is also expressed in other locations, notably within the spermatogonial stem/progenitor cell population in postnatal testis. Independent studies have shown that Sox3 null mice exhibit a spermatogenic block as young adults, the mechanism of which remains poorly understood. Using a panel of spermatogonial cell marker genes, we demonstrate that Sox3 is expressed within the committed progenitor fraction of the undifferentiated spermatogonial pool. Additionally, we use a Sox3 null mouse model to define a potential role for this factor in progenitor cell function. We demonstrate that Sox3 expression is required for transition of undifferentiated cells from a GFRα1+ self-renewing state to the NGN3 + transit-amplifying compartment. Critically, using chromatin immunoprecipitation, we demonstrate that SOX3 binds to a highly conserved region in the Ngn3 promoter region in vivo, indicating that Ngn3 is a direct target of SOX3. Together these studies indicate that SOX3 functions as a pro-commitment factor in spermatogonial stem/progenitor cells.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , Factores de Transcripción SOXB1/genética , Espermatogonias/metabolismo , Testículo/metabolismo , Células Madre Germinales Adultas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Unión Proteica , Factores de Transcripción SOXB1/deficiencia , Transducción de Señal , Espermatogénesis/genética , Espermatogonias/citología , Espermatogonias/crecimiento & desarrollo , Testículo/citología , Testículo/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Biol Open ; 9(2)2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31988094

RESUMEN

In the anterior foregut (AFG) of mouse embryos, the transcription factor SOX2 is expressed in the epithelia of the esophagus and proximal branches of respiratory organs comprising the trachea and bronchi, whereas NKX2.1 is expressed only in the epithelia of respiratory organs. Previous studies using hypomorphic Sox2 alleles have indicated that reduced SOX2 expression causes the esophageal epithelium to display some respiratory organ characteristics. In the present study, we produced mouse embryos with AFG-specific SOX2 deficiency. In the absence of SOX2 expression, a single NKX2.1-expressing epithelial tube connected the pharynx and the stomach, and a pair of bronchi developed in the middle of the tube. Expression patterns of NKX2.1 and SOX9 revealed that the anterior and posterior halves of SOX2-deficient AFG epithelial tubes assumed the characteristics of the trachea and bronchus, respectively. In addition, we found that mesenchymal tissues surrounding the SOX2-deficient NKX2.1-expressing epithelial tube changed to those surrounding the trachea and bronchi in the anterior and posterior halves, as indicated by the arrangement of smooth muscle cells and SOX9-expressing cells and by the expression of Wnt4 (esophagus specific), Tbx4 (respiratory organ specific), and Hoxb6 (distal bronchus specific). The impact of mesenchyme-derived signaling on the early stage of AFG epithelial specification has been indicated. Our study demonstrated an opposite trend where epithelial tissue specification causes concordant changes in mesenchymal tissues, indicating a reciprocity of epithelial-mesenchymal interactions.


Asunto(s)
Esófago/anomalías , Tracto Gastrointestinal/anomalías , Organogénesis/genética , Factores de Transcripción SOXB1/deficiencia , Tráquea/anomalías , Animales , Diferenciación Celular/genética , Endodermo/anomalías , Endodermo/embriología , Epitelio/embriología , Esófago/embriología , Técnica del Anticuerpo Fluorescente , Tracto Gastrointestinal/embriología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/embriología , Ratones , Ratones Transgénicos , Tráquea/embriología
5.
Cell Mol Life Sci ; 77(18): 3597-3609, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31758234

RESUMEN

The bHLH transcription factor Olig2 is required for sequential cell fate determination of both motor neurons and oligodendrocytes and for progenitor proliferation in the central nervous system. However, the role of Olig2 in peripheral sensory neurogenesis remains unknown. We report that Olig2 is transiently expressed in the newly differentiated olfactory sensory neurons (OSNs) and is down-regulated in the mature OSNs in mice from early gestation to adulthood. Genetic fate mapping demonstrates that Olig2-expressing cells solely give rise to OSNs in the peripheral olfactory system. Olig2 depletion does not affect the proliferation of peripheral olfactory progenitors and the fate determination of OSNs, sustentacular cells, and the olfactory ensheathing cells. However, the terminal differentiation and maturation of OSNs are compromised in either Olig2 single or Olig1/Olig2 double knockout mice, associated with significantly diminished expression of multiple OSN maturation and odorant signaling genes, including Omp, Gnal, Adcy3, and Olfr15. We further demonstrate that Olig2 binds to the E-box in the Omp promoter region to regulate its expression. Taken together, our results reveal a distinctly novel function of Olig2 in the periphery nervous system to regulate the terminal differentiation and maturation of olfactory sensory neurons.


Asunto(s)
Diferenciación Celular , Neuronas Receptoras Olfatorias/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linaje de la Célula , Proliferación Celular , Proteína Doblecortina , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Ratones , Ratones Transgénicos , Proteína Marcadora Olfativa/genética , Mucosa Olfatoria/citología , Mucosa Olfatoria/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/deficiencia , Factor de Transcripción 2 de los Oligodendrocitos/genética , Regiones Promotoras Genéticas , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
6.
Cell Stem Cell ; 24(3): 462-476.e6, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30849367

RESUMEN

The SOX2 transcription factor is critical for neural stem cell (NSC) maintenance and brain development. Through chromatin immunoprecipitation (ChIP) and chromatin interaction analysis (ChIA-PET), we determined genome-wide SOX2-bound regions and Pol II-mediated long-range chromatin interactions in brain-derived NSCs. SOX2-bound DNA was highly enriched in distal chromatin regions interacting with promoters and carrying epigenetic enhancer marks. Sox2 deletion caused widespread reduction of Pol II-mediated long-range interactions and decreased gene expression. Genes showing reduced expression in Sox2-deleted cells were significantly enriched in interactions between promoters and SOX2-bound distal enhancers. Expression of one such gene, Suppressor of Cytokine Signaling 3 (Socs3), rescued the self-renewal defect of Sox2-ablated NSCs. Our work identifies SOX2 as a major regulator of gene expression through connections to the enhancer network in NSCs. Through the definition of such a connectivity network, our study shows the way to the identification of genes and enhancers involved in NSC maintenance and neurodevelopmental disorders.


Asunto(s)
Cromatina/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Células Cultivadas , Redes Reguladoras de Genes/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Pez Cebra
7.
Theranostics ; 9(2): 424-435, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809284

RESUMEN

Tumorigenic cells, when facing a hostile environment, may enter a dormant state, leading to long-term tumor survival, relapse, and metastasis. To date, the molecular mechanism of tumor cell dormancy remains poorly understood. Methods: A soft, 3-dimentional (3D) fibrin gel culture system was used to mechanically select and grow highly malignant and tumorigenic melanoma tumor-repopulating cells (TRCs). We cultured control melanoma TRCs, TRCs with Sox2 knockdown, TRCs with Sox2 knockout, and a 2D control for in vitro and in vivo experiments. Western blotting, immunofluorescence, and flow cytometry analysis were performed to examine TRC dormancy and exit from dormancy. Results: Under a low-expression condition, we show that Sox2, a stemness molecule participates in dormancy regulation of highly tumorigenic cells that can repopulate a tumor (TRCs). Intriguingly, complete depletion of Sox2 via knockout results in dormancy exit and growth resumption of melanoma TRCs in culture and elevation of melanoma TRC apoptosis. Mice that are injected subcutaneously with Sox2-depleted melanoma TRCs do not form tumors and survive much longer than those injected with melanoma TRCs. We found that complete depletion of Sox2 promotes nuclear translocation of phosphorylated STAT3, where it binds to the p53 gene promoter, thus activating the p53-caspase3 cascade. Conclusion: These findings provide a novel insight into the role of the Sox2 gene in tumor cell stemness, tumor dormancy, and apoptosis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma Experimental/patología , Células Madre Neoplásicas/patología , Factores de Transcripción SOXB1/metabolismo , Animales , Apoptosis , Proliferación Celular , Ratones , Recurrencia Local de Neoplasia , Factores de Transcripción SOXB1/deficiencia
8.
Cereb Cortex ; 29(1): 54-69, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29161339

RESUMEN

Injury to the adult brain induces activation of local astrocytes, which serves as a compensatory response that modulates tissue damage and recovery. However, the mechanism governing astrocyte activation during brain injury remains largely unknown. Here we provide in vivo evidence that SOX2, a transcription factor critical for stem cells and brain development, is also required for injury-induced activation of adult cortical astrocytes. Genome-wide chromatin immunoprecipitation-seq analysis of mouse cortical tissues reveals that SOX2 binds to regulatory regions of genes associated with signaling pathways that control glial cell activation, such as Nr2e1, Mmd2, Wnt7a, and Akt2. Astrocyte-specific deletion of Sox2 in adult mice greatly diminishes glial response to controlled cortical impact injury and, most unexpectedly, dampens injury-induced cortical loss and benefits behavioral recovery of mice after injury. Together, these results uncover an essential role of SOX2 in somatic cells under pathological conditions and indicate that SOX2-dependent astrocyte activation could be targeted for functional recovery after traumatic brain injury.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Eliminación de Gen , Recuperación de la Función/fisiología , Factores de Transcripción SOXB1/deficiencia , Animales , Astrocitos/patología , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Células Cultivadas , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales , Factores de Transcripción SOXB1/genética
9.
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
10.
Nature ; 548(7665): 52-57, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28746310

RESUMEN

It has been proposed that the hypothalamus helps to control ageing, but the mechanisms responsible remain unclear. Here we develop several mouse models in which hypothalamic stem/progenitor cells that co-express Sox2 and Bmi1 are ablated, as we observed that ageing in mice started with a substantial loss of these hypothalamic cells. Each mouse model consistently displayed acceleration of ageing-like physiological changes or a shortened lifespan. Conversely, ageing retardation and lifespan extension were achieved in mid-aged mice that were locally implanted with healthy hypothalamic stem/progenitor cells that had been genetically engineered to survive in the ageing-related hypothalamic inflammatory microenvironment. Mechanistically, hypothalamic stem/progenitor cells contributed greatly to exosomal microRNAs (miRNAs) in the cerebrospinal fluid, and these exosomal miRNAs declined during ageing, whereas central treatment with healthy hypothalamic stem/progenitor cell-secreted exosomes led to the slowing of ageing. In conclusion, ageing speed is substantially controlled by hypothalamic stem cells, partially through the release of exosomal miRNAs.


Asunto(s)
Envejecimiento/genética , Envejecimiento/fisiología , Exosomas/genética , Hipotálamo/citología , Hipotálamo/fisiología , Longevidad/fisiología , MicroARNs/genética , Células-Madre Neurales/fisiología , Envejecimiento/líquido cefalorraquídeo , Envejecimiento/patología , Animales , Microambiente Celular , Exosomas/metabolismo , Hipotálamo/patología , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamación , Longevidad/genética , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/líquido cefalorraquídeo , MicroARNs/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Complejo Represivo Polycomb 1/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Factores de Transcripción SOXB1/deficiencia , Factores de Tiempo
11.
Oncotarget ; 7(30): 47095-47110, 2016 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-27283990

RESUMEN

Type II germ cell cancers (GCC) are divided into seminomas, which are highly similar to primordial germ cells and embryonal carcinomas (EC), often described as malignant counterparts to embryonic stem cells.Previously, we demonstrated that the development of GCCs is a highly plastic process and strongly influenced by the microenvironment. While orthotopic transplantation into the testis promotes seminomatous growth of the seminoma-like cell line TCam-2, ectopic xenotransplantation into the flank initiates reprogramming into an EC-like fate.During this reprogramming, BMP signaling is inhibited, leading to induction of NODAL signaling, upregulation of pluripotency factors and downregulation of seminoma markers, like SOX17. The pluripotency factor and EC-marker SOX2 is strongly induced.Here, we adressed the molecular role of SOX2 in this reprogramming. Using CRISPR/Cas9-mediated genome-editing, we established SOX2-deficient TCam-2 cells. Xenografting of SOX2-deficient cells into the flank of nude mice resulted in maintenance of a seminoma-like fate, indicated by the histology and expression of OCT3/4, SOX17, TFAP2C, PRDM1 and PRAME. In SOX2-deficient cells, BMP signaling is inhibited, but NODAL signaling is not activated. Thus, SOX2 appears to be downstream of BMP signaling but upstream of NODAL activation. So, SOX2 is an essential factor in acquiring an EC-like cell fate from seminomas.A small population of differentiated cells was identified resembling a mixed non-seminoma. Analyses of these cells revealed downregulation of the pluripotency and seminoma markers OCT3/4, SOX17, PRDM1 and TFAP2C. In contrast, the pioneer factor FOXA2 and its target genes were upregulated, suggesting that FOXA2 might play an important role in induction of non-seminomatous differentiation.


Asunto(s)
Carcinoma Embrionario/patología , Factores de Transcripción SOXB1/metabolismo , Seminoma/patología , Animales , Carcinoma Embrionario/genética , Carcinoma Embrionario/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Reprogramación Celular/fisiología , Técnicas de Inactivación de Genes , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Proteína Nodal/metabolismo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Seminoma/genética , Seminoma/metabolismo , Transfección
12.
J Histochem Cytochem ; 63(9): 721-33, 2015 09.
Artículo en Inglés | MEDLINE | ID: mdl-26001828

RESUMEN

SRY (sex determining region Y)-box 2 (SOX2) plays an important role in tumor cell metastasis and apoptosis. Laryngeal squamous cell carcinoma (LSCC), responsible for 1.5% of all cancers, is one of the most common head and neck malignancies. Accumulating evidence shows that SOX2 is overexpressed in several human tumors, including lung cancer, esophageal carcinoma, pancreatic carcinoma, breast cancer, ovarian carcinoma and glioma. Our study aimed to investigate the silencing effects of SOX2 expression using RNA interference (RNAi) on various biological processes in laryngeal cancer TU212 cells, including proliferation, apoptosis, invasion and metastasis. We also studied the involvement of the MAPK/JNK signaling pathway in the biological effects of SOX2 siRNA in TU212 cells. We found that silencing SOX2 decreased the proliferation, migration, and invasion of TU212 cells, and induced apoptosis. This effect of silencing SOX2 could be reversed by silencing MAP4K4. Therefore, we consider SOX2 as a key regulator of the upstream MAP4K4/JNK signaling pathways that could be a potential therapeutic target in the treatment of patients with or prevention of laryngeal cancer.


Asunto(s)
Apoptosis/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Laríngeas/patología , Sistema de Señalización de MAP Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Factores de Transcripción SOXB1/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Células Clonales/patología , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Activación Enzimática/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Laríngeas/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación/genética , ARN Interferente Pequeño/genética , Factores de Transcripción SOXB1/deficiencia
13.
Proc Natl Acad Sci U S A ; 112(15): E1936-45, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825708

RESUMEN

Newborn granule neurons generated from neural progenitor cells (NPCs) in the adult hippocampus play a key role in spatial learning and pattern separation. However, the molecular mechanisms that control activation of their neurogenic program remain poorly understood. Here, we report a novel function for the pluripotency factor sex-determining region Y (SRY)-related HMG box 2 (SOX2) in regulating the epigenetic landscape of poised genes activated at the onset of neuronal differentiation. We found that SOX2 binds to bivalently marked promoters of poised proneural genes [neurogenin 2 (Ngn2) and neurogenic differentiation 1 (NeuroD1)] and a subset of neurogenic genes [e.g., SRY-box 21 (Sox21), brain-derived neurotrophic factor (Bdnf), and growth arrest and DNA-damage-inducible, beta (Gadd45b)] where it functions to maintain the bivalent chromatin state by preventing excessive polycomb repressive complex 2 activity. Conditional ablation of SOX2 in adult hippocampal NPCs impaired the activation of proneural and neurogenic genes, resulting in increased neuroblast death and functionally aberrant newborn neurons. We propose that SOX2 sets a permissive epigenetic state in NPCs, thus enabling proper activation of the neuronal differentiation program under neurogenic cue.


Asunto(s)
Epigénesis Genética , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Factores de Transcripción SOXB1/genética , Activación Transcripcional , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Ciclo Celular/genética , Proliferación Celular/genética , Células Cultivadas , Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Técnicas de Placa-Clamp , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/metabolismo
14.
PLoS One ; 10(2): e0116839, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25642713

RESUMEN

Melanoma tissues and cell lines are heterogeneous, and include cells with invasive, proliferative, stem cell-like, and differentiated properties. Such heterogeneity likely contributes to the aggressiveness of the disease and resistance to therapy. One model suggests that heterogeneity arises from rare cancer stem cells (CSCs) that produce distinct cancer cell lineages. Another model suggests that heterogeneity arises through reversible cellular plasticity, or phenotype-switching. Recent work indicates that phenotype-switching may include the ability of cancer cells to dedifferentiate to a stem cell-like state. We set out to investigate the phenotype-switching capabilities of melanoma cells, and used unbiased methods to identify genes that may control such switching. We developed a system to reversibly synchronize melanoma cells between 2D-monolayer and 3D-stem cell-like growth states. Melanoma cells maintained in the stem cell-like state showed a striking upregulation of a gene set related to development and neural stem cell biology, which included SRY-box 2 (SOX2) and Inhibitor of DNA Binding 4 (ID4). A gene set related to cancer cell motility and invasiveness was concomitantly downregulated. Intense and pervasive ID4 protein expression was detected in human melanoma tissue samples, suggesting disease relevance for this protein. SiRNA knockdown of ID4 inhibited switching from monolayer to 3D-stem cell-like growth, and instead promoted switching to a highly differentiated, neuronal-like morphology. We suggest that ID4 is upregulated in melanoma as part of a stem cell-like program that facilitates further adaptive plasticity. ID4 may contribute to disease by preventing stem cell-like melanoma cells from progressing to a normal differentiated state. This interpretation is guided by the known role of ID4 as a differentiation inhibitor during normal development. The melanoma stem cell-like state may be protected by factors such as ID4, thereby potentially identifying a new therapeutic vulnerability to drive differentiation to the normal cell phenotype.


Asunto(s)
Diferenciación Celular , Regulación Neoplásica de la Expresión Génica , Proteínas Inhibidoras de la Diferenciación/metabolismo , Melanoma/metabolismo , Melanoma/patología , Adhesión Celular , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Inhibidoras de la Diferenciación/deficiencia , Proteínas Inhibidoras de la Diferenciación/genética , Melanoma/genética , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Fenotipo , ARN Interferente Pequeño/genética , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Transcripción Genética
15.
Oncotarget ; 6(5): 2992-3002, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25576924

RESUMEN

Cellular reprogramming to iPSCs has uncovered unsuspected links between tumor suppressors and pluripotency factors. Using this system, it was possible to identify tumor suppressor p27 as a repressor of Sox2 during differentiation. This led to the demonstration that defects in the repression of Sox2 can contribute to tumor development. The members of the retinoblastoma family of pocket proteins, pRb, p107 and p130, are negative regulators of the cell cycle with tumor suppressor activity and with roles in differentiation. In this work we studied the relative contribution of the retinoblastoma family members to the regulation of Sox2 expression. We found that deletion of Rb or p130 leads to impaired repression of Sox2, a deffect amplified by inactivation of p53. We also identified binding of pRb and p130 to an enhancer with crucial regulatory activity on Sox2 expression. Using cellular reprogramming we tested the impact of the defective repression of Sox2 and confirmed that Rb deficiency allows the generation of iPSCs in the absence of exogenous Sox2. Finally, partial depletion of Sox2 positive cells reduced the pituitary tumor development initiated by Rb loss in vivo. In summary, our results show that Sox2 repression by pRb is a relevant mechanism of tumor suppression.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transcripción Genética , Animales , Reprogramación Celular , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genotipo , Células HEK293 , Humanos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Fenotipo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Interferencia de ARN , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/deficiencia , Proteína p130 Similar a la del Retinoblastoma/genética , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Transfección , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
16.
Cleft Palate Craniofac J ; 51(1): 110-4, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23638914

RESUMEN

OBJECTIVE: While SEX-determining region Y-Box 2 (SOX2) mutations are typically recognized as yielding ocular and central nervous system abnormalities, they have also been associated with other craniofacial defects. To elucidate the genesis of the latter, Sox2 hypomorphic (Sox2(HYP)) mice were examined, with particular attention to secondary palatal development. RESULTS: Clefts of the secondary palate were found to be highly penetrant in Sox2(HYP) mice. The palatal clefting occurred in the absence of mandibular hypoplasia and resulted from delayed or failed shelf elevation. CONCLUSIONS: Sox2 hypomorphism can result in clefting of the secondary palate, an effect that appears to be independent of mandibular hypoplasia and is thus expected to result from an abnormality that is inherent to the palatal shelves and/or their progenitor tissues. Further clinical attention relative to SOX2 mutations as a basis for secondary palatal clefts appears warranted.


Asunto(s)
Fisura del Paladar/genética , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Alelos , Animales , Modelos Animales de Enfermedad , Genotipo , Ratones
17.
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
18.
Stem Cells Dev ; 22(12): 1779-88, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23316968

RESUMEN

Skin-derived precursors (SKPs) are an attractive stem cell model for cell-based therapies. SKPs can be readily generated from embryonic and adult mice and adult humans, exhibit a high degree of multipotency, and have the potential to serve as a patient autologous stem cell. The advancement of these cells toward therapeutic use depends on the ability to control precisely the self-renewal and differentiation of SKPs. Here we show that two well-known stem cell factors, Foxd3 and Sox2, are critical regulators of the stem cell properties of SKPs. Deletion of Foxd3 completely abolishes the sphere-forming potential of these cells. In the absence of Sox2, SKP spheres can be formed, but with reduced size and frequency. Our results provide entry points into the gene regulatory networks dictating SKP behavior, and pave the way for future studies on a therapeutically relevant stem cell.


Asunto(s)
Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Multipotentes/metabolismo , Proteínas Represoras/genética , Factores de Transcripción SOXB1/genética , Piel/metabolismo , Transcripción Genética , Animales , Diferenciación Celular , Embrión de Mamíferos , Células Madre Embrionarias/citología , Factores de Transcripción Forkhead/deficiencia , Ratones , Ratones Transgénicos , Células Madre Multipotentes/citología , Cultivo Primario de Células , Proteínas Represoras/deficiencia , Factores de Transcripción SOXB1/deficiencia , Transducción de Señal , Piel/citología , Piel/embriología
19.
Dev Cell ; 23(5): 981-94, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23153495

RESUMEN

How dermal papilla (DP) niche cells regulate hair follicle progenitors to control hair growth remains unclear. Using Tbx18(Cre) to target embryonic DP precursors, we ablate the transcription factor Sox2 early and efficiently, resulting in diminished hair shaft outgrowth. We find that DP niche expression of Sox2 controls the migration speed of differentiating hair shaft progenitors. Transcriptional profiling of Sox2 null DPs reveals increased Bmp6 and decreased BMP inhibitor Sostdc1, a direct Sox2 transcriptional target. Subsequently, we identify upregulated BMP signaling in knockout hair shaft progenitors and demonstrate that Bmp6 inhibits cell migration, an effect that can be attenuated by Sostdc1. A shorter and Sox2-negative hair type lacks Sostdc1 in the DP and shows reduced migration and increased BMP activity of hair shaft progenitors. Collectively, our data identify Sox2 as a key regulator of hair growth that controls progenitor migration by fine-tuning BMP-mediated mesenchymal-epithelial crosstalk.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Folículo Piloso/embriología , Folículo Piloso/metabolismo , Cabello/crecimiento & desarrollo , Factores de Transcripción SOXB1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteína Morfogenética Ósea 6/metabolismo , Proteínas Morfogenéticas Óseas/deficiencia , Movimiento Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Folículo Piloso/citología , Folículo Piloso/crecimiento & desarrollo , Ratones , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Transducción de Señal , Transcriptoma
20.
J Clin Invest ; 122(10): 3635-46, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22945632

RESUMEN

Sex-determining region Y (SRY) box 2 (SOX2) haploinsufficiency causes a form of hypopituitarism in humans that is characterized by gonadotrophin deficiency known as hypogonadotrophic hypogonadism. Here, we conditionally deleted Sox2 in mice to investigate the pathogenesis of hypogonadotrophic hypogonadism. First, we found that absence of SOX2 in the developing Rathke pouch of conditional embryos led to severe anterior lobe hypoplasia with drastically reduced expression of the pituitary-specific transcription factor POU class 1 homeobox 1 (POU1F1) as well as severe disruption of somatotroph and thyrotroph differentiation. In contrast, corticotrophs, rostral-tip POU1F1-independent thyrotrophs, and, interestingly, lactotrophs and gonadotrophs were less affected. Second, we identified a requirement for SOX2 in normal proliferation of periluminal progenitors; in its absence, insufficient precursors were available to produce all cell lineages of the anterior pituitary. Differentiated cells derived from precursors exiting cell cycle at early stages, including corticotrophs, rostral-tip thyrotrophs, and gonadotrophs, were generated, while hormone-producing cells originating from late-born precursors, such as somatotrophs and POU1F1-dependent thyrotrophs, were severely reduced. Finally, we found that 2 previously characterized patients with SOX2 haploinsufficiency and associated hypogonadotrophic hypogonadism had a measurable response to gonadotropin-releasing hormone (GnRH) stimulation, suggesting that it is not the absence of gonadotroph differentiation, but rather the deficient hypothalamic stimulation of gonadotrophs, that underlies typical hypogonadotrophic hypogonadism.


Asunto(s)
Hipogonadismo/genética , Sistema Hipotálamo-Hipofisario/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Femenino , Regulación del Desarrollo de la Expresión Génica , Hormona Liberadora de Gonadotropina/uso terapéutico , Heterocigoto , Proteínas de Homeodominio/genética , Humanos , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/fisiopatología , Ratones , Ratones Noqueados , Organogénesis/genética , Organogénesis/fisiología , Adenohipófisis/anomalías , Adenohipófisis/embriología , Adenohipófisis/metabolismo , Adenohipófisis/patología , Proteínas Represoras/genética , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Somatotrofos/patología , Tirotrofos/patología , Factor de Transcripción Pit-1/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...