Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Sci ; 112(10): 4176-4186, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34327778

RESUMEN

As a POU homeodomain transcription factor, POU4F2 has been implicated in regulating tumorigenic processes in various cancers. However, the role of POU4F2 in colorectal cancer (CRC) remains unclear. Here, we revealed that POU4F2 functions as a tumor promotor in CRC. Bioinformatics analysis in specimens from CRC patients and expression analysis in CRC cell lines showed that POU4F2 was upregulated at the mRNA and protein levels in CRC. Depletion of POU4F2 suppressed the metastatic phenotypes of CRC cells, including cell migration, invasion, and the expression of epithelial-mesenchymal transition (EMT) markers. Moreover, depletion of POU4F2 decreased the number of lung metastatic nodes in nude mice. Mechanistically, POU4F2 positively regulated the Hedgehog signaling pathway, as inferred from the downregulation of the expression of sonic Hedgehog homolog, patched 1, Smoothened, and GLI family zinc finger 1 in vitro and vivo following silencing of POU4F2. Furthermore, the SMO agonist SAG reversed the effects of POU4F2 knockdown in CRC. Functionally, POU4F2 contributed to the Hedgehog signaling-regulated activation of the EMT process and promotion of CRC cell migration and invasion. Collectively, these findings elucidated the role of POU4F2 as a tumor promotor in CRC through the regulation of Hedgehog signaling-mediated EMT and suggested that POU4F2 suppression might be a promising therapeutic target in inhibiting CRC metastasis.


Asunto(s)
Movimiento Celular , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Proteínas Hedgehog/metabolismo , Invasividad Neoplásica , Factor de Transcripción Brn-3B/fisiología , Animales , Línea Celular Tumoral , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/patología , Ciclohexilaminas/farmacología , Regulación hacia Abajo , Silenciador del Gen , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Terapia Molecular Dirigida , Receptor Patched-1/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Receptor Smoothened/agonistas , Receptor Smoothened/metabolismo , Tiofenos/farmacología , Factor de Transcripción Brn-3B/antagonistas & inhibidores , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo , Regulación hacia Arriba , Dedos de Zinc
2.
Stem Cell Reports ; 15(5): 1095-1110, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33096050

RESUMEN

Direct reprogramming has been widely explored to generate various types of neurons for neurobiological research and translational medicine applications, but there is still no efficient reprogramming method to generate retinal ganglion cell (RGC)-like neurons, which are the sole projection neurons in the retina. Here, we show that three transcription factors, Ascl1, Brn3b, and Isl1, efficiently convert fibroblasts into RGC-like neurons (iRGCs). Furthermore, we show that the competence of cells to enter iRGC reprogramming route is determined by the cell-cycle status at a very early stage of the process. The iRGC reprogramming route involves intermediate states that are characterized by a transient inflammatory-like response followed by active epigenomic and transcriptional modifications. Our study provides an efficient method to generate iRGCs, which would be a valuable cell source for potential glaucoma cell replacement therapy and drug screening studies, and reveals the key cellular events that govern successful neuronal fate reprogramming.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Reprogramación Celular , Fibroblastos/fisiología , Proteínas de Homeodominio/fisiología , Proteínas con Homeodominio LIM/fisiología , Neuronas/fisiología , Células Ganglionares de la Retina/fisiología , Factor de Transcripción Brn-3B/fisiología , Factores de Transcripción/fisiología , Animales , Ciclo Celular , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Neurogénesis , Retina/citología
3.
J Neurophysiol ; 124(5): 1530-1549, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32997561

RESUMEN

A variety of visual cues can trigger defensive reactions in mice and other species. In mice, looming stimuli that mimic an approaching aerial predator elicit flight or freezing reactions, while sweeping stimuli that mimic an aerial predator flying parallel to the ground typically elicit freezing. The retinal ganglion cell (RGC) types involved in these circuits are largely unknown. We previously discovered that loss of RGC subpopulations in Brn3b knockout mice results in distinct visual response deficits. Here, we report that retinal or global loss of Brn3b selectively ablates the fleeing response to looming stimuli while leaving the freeze response intact. In contrast, freezing responses to sweeping stimuli are significantly affected. Genetic manipulations removing three RGC subpopulations (Brn3a+ betta RGCs, Opn4+Brn3b+, and Brn3c+Brn3b+ RGCs) result in milder phenocopies of Brn3b knockout response deficits. These findings show that flight and freezing responses to distinct visual cues are mediated by circuits that can already be separated at the level of the retina, potentially by enlisting dedicated RGC types.NEW & NOTEWORTHY Flight and freezing response choices evoked by visual stimuli are controlled by brain stem and thalamic circuits. Genetically modified mice with loss of specific retinal ganglion cell (RGC) subpopulations have altered flight versus freezing choices in response to some but not other visual stimuli. This finding suggests that "threatening" visual stimuli may be computed already at the level of the retina and communicated via dedicated pathways (RGCs) to the brain.


Asunto(s)
Reacción de Prevención/fisiología , Células Ganglionares de la Retina/fisiología , Percepción Visual/fisiología , Animales , Conducta Animal , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/fisiología
4.
Dev Biol ; 364(2): 114-27, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22326227

RESUMEN

The sensory neurons of the dorsal root ganglia (DRG) must project accurately to their central targets to convey proprioceptive, nociceptive and mechanoreceptive information to the spinal cord. How these different sensory modalities and central connectivities are specified and coordinated still remains unclear. Given the expression of the POU homeodomain transcription factors Brn3a/Pou4f1 and Brn3b/Pou4f2 in DRG and spinal cord sensory neurons, we determined the subtype specification of DRG and spinal cord sensory neurons as well as DRG central projections in Brn3a and Brn3b single and double mutant mice. Inactivation of either or both genes causes no gross abnormalities in early spinal cord neurogenesis; however, in Brn3a single and Brn3a;Brn3b double mutant mice, sensory afferent axons from the DRG fail to form normal trajectories in the spinal cord. The TrkA(+) afferents remain outside the dorsal horn and fail to extend into the spinal cord, while the projections of TrkC(+) proprioceptive afferents into the ventral horn are also impaired. Moreover, Brn3a mutant DRGs are defective in sensory neuron specification, as marked by the excessive generation of TrkB(+) and TrkC(+) neurons as well as TrkA(+)/TrkB(+) and TrkA(+)/TrkC(+) double positive cells at early embryonic stages. At later stages in the mutant, TrkB(+), TrkC(+) and parvalbumin(+) neurons diminish while there is a significant increase of CGRP(+) and c-ret(+) neurons. In addition, Brn3a mutant DRGs display a dramatic down-regulation of Runx1 expression, suggesting that the regulation of DRG sensory neuron specification by Brn3a is mediated in part by Runx1. Our results together demonstrate a critical role for Brn3a in generating DRG sensory neuron diversity and regulating sensory afferent projections to the central targets.


Asunto(s)
Axones/fisiología , Ganglios Espinales/embriología , Médula Espinal/embriología , Factor de Transcripción Brn-3A/fisiología , Animales , Péptido Relacionado con Gen de Calcitonina/análisis , Subunidad alfa 2 del Factor de Unión al Sitio Principal/análisis , Regulación hacia Abajo , Femenino , Ganglios Espinales/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Masculino , Ratones , Neurogénesis/fisiología , Proteínas Proto-Oncogénicas c-ret/análisis , Receptor trkA/análisis , Receptor trkB/análisis , Receptor trkC/análisis , Médula Espinal/crecimiento & desarrollo , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/fisiología
5.
Dev Dyn ; 238(9): 2309-17, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19459208

RESUMEN

Although immunological detection of proteins is used extensively in retinal development, studies are often impeded because antibodies against crucial proteins cannot be generated or are not readily available. Here, we overcome these limitations by constructing genetically engineered alleles for Math5 and Pou4f2, two genes required for retinal ganglion cell (RGC) development. Sequences encoding a peptide epitope from haemagglutinin (HA) were added to Math5 or Pou4f2 in frame to generate Math5(HA) and Pou4f2(HA) alleles. We demonstrate that the tagged alleles recapitulated the wild-type expression patterns of the two genes, and that the tags did not interfere with the function of the cognate proteins. In addition, by co-staining, we found that Math5 and Pou4f2 were transiently co-expressed in newly born RGCs, unequivocally demonstrating that Pou4f2 is immediately downstream of Math5 in RGC formation. The epitope-tagged alleles provide new and useful tools for analyzing gene regulatory networks underlying RGC development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Epítopos/genética , Proteínas de Homeodominio/fisiología , Proteínas del Tejido Nervioso/fisiología , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Southern Blotting , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo
6.
Development ; 135(11): 1981-90, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18434421

RESUMEN

LIM-homeodomain (HD) and POU-HD transcription factors play crucial roles in neurogenesis. However, it remains largely unknown how they cooperate in this process and what downstream target genes they regulate. Here, we show that ISL1, a LIM-HD protein, is co-expressed with BRN3B, a POU-HD factor, in nascent post-mitotic retinal ganglion cells (RGCs). Similar to the Brn3b-null retinas, retina-specific deletion of Isl1 results in the apoptosis of a majority of RGCs and in RGC axon guidance defects. The Isl1 and Brn3b double null mice display more severe retinal abnormalities with a near complete loss of RGCs, indicating the synergistic functions of these two factors. Furthermore, we show that both Isl1 and Brn3b function downstream of Math5 to regulate the expression of a common set of RGC-specific genes. Whole-retina chromatin immunoprecipitation and in vitro transactivation assays reveal that ISL1 and BRN3B concurrently bind to and synergistically regulate the expression of a common set of RGC-specific genes. Thus, our results uncover a novel regulatory mechanism of BRN3B and ISL1 in RGC differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Homeodominio/fisiología , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/fisiología , Animales , Diferenciación Celular/genética , Inmunoprecipitación de Cromatina , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Proteínas con Homeodominio LIM , Ratones , Ratones Noqueados , Mitosis , Regiones Promotoras Genéticas/genética , Unión Proteica , Retina/citología , Retina/embriología , Retina/metabolismo , Células Ganglionares de la Retina/citología , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo , Factores de Transcripción
7.
J Neurosci ; 28(13): 3392-403, 2008 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-18367606

RESUMEN

The retinal ganglion cells (RGCs) are the sole output neurons in the retina that form the optic nerve and convey light signals detected by photoreceptors to the higher visual system. Their degeneration and damage caused by glaucoma and injury can lead to blindness. During retinogenesis, RGCs are specified from a population of multipotential precursors capable of generating RGC, amacrine, horizontal, and cone cells. How the RGC fate is selected from these multiple neuron fates is unknown at present. Here we show that the previously unsuspected POU domain transcription factor Brn3b (brain-specific homeobox/POU domain protein 3b) plays such a critical role. Loss of Brn3b function in mice leads to misspecification of early RGC precursors as late-born RGC, amacrine, and horizontal cells, whereas misexpressed Brn3b suppresses non-RGC cell fates but promotes the RGC fate. Microarray profiling and other molecular analyses reveal that, in RGC precursors, Brn3b normally represses the expression of a network of retinogenic factor genes involved in fate commitment and differentiation of late-born RGC, amacrine, horizontal, and cone cells. Our data suggest that Brn3b specifies the RGC fate from multipotential precursors not only by promoting RGC differentiation but also by suppressing non-RGC differentiation programs as a safeguard mechanism.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/fisiología , Retina/citología , Células Ganglionares de la Retina/fisiología , Factor de Transcripción Brn-3B/fisiología , Células Amacrinas/metabolismo , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Diferenciación Celular/genética , Análisis por Conglomerados , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Operón Lac/fisiología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Células Fotorreceptoras/embriología , Factor de Transcripción Brn-3B/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Oncogene ; 27(1): 145-54, 2008 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-17637757

RESUMEN

Brn-3b transcription factor enhances proliferation of neuroblastoma (NB) and breast cancer cell lines in vitro and increases the rate and size of in vivo tumour growth, whereas reducing Brn-3b slows growth, both in vitro and in vivo. Brn-3b is elevated in >65% of breast cancer biopsies, and here we demonstrate that Brn-3b is also elevated in NB tumours. We show a significant correlation between Brn-3b and cyclin D1 (CD1) in breast cancers and NB tumours and cell lines. Brn-3b directly transactivates the CD1 promoter in co-transfection experiments, whereas electrophoretic mobility shift assay and chromatin immunoprecipitation assays demonstrate that Brn-3b protein binds to an octamer sequence located in the proximal CD1 promoter. Site-directed mutagenesis of this sequence resulted in loss of transactivation of the CD1 promoter by Brn-3b. Thus, Brn-3b may act to alter growth properties of breast cancer and NB cells by enhancing CD1 expression in these cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica , Neuroblastoma/metabolismo , Factor de Transcripción Brn-3B/fisiología , Activación Transcripcional , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ciclina D1/biosíntesis , Femenino , Humanos , Neuroblastoma/patología , ARN Mensajero/biosíntesis , ARN Mensajero/metabolismo , Factor de Transcripción Brn-3B/biosíntesis , Factor de Transcripción Brn-3B/genética , Células Tumorales Cultivadas , Regulación hacia Arriba/fisiología
9.
J Cell Biochem ; 102(1): 1-12, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17668438

RESUMEN

Osteoclastic bone resorption is a central mechanism in skeletal development, remodeling and pathology. RANKL is a mandatory factor controlling osteoclastogenesis; however, the underlying signaling pathways are only partially characterized. Using a screening array for the investigation of differential transcription factor activation, we identified activation of the Brn3 transcription factor family as a downstream event of RANKL signaling during terminal osteoclastogenesis. RANKL stimulation induces expression of Brn3a and b and maximal transcriptional activity of Brn3 family members concurrent with osteoclastic giant cell formation. Immunohistochemical analysis revealed both nuclear and cytoplasmic localization of Brn3a and b in mature osteoclasts. Functional inhibition of Brn3 transcription factors resulted in inhibition of pre-osteoclast fusion and reduction in bone resorbing activity of mature osteoclasts. Furthermore, we identified synaptotagmin-1, a regulator of membrane and vesicular fusion, as downstream target of Brn3 with a role in osteoclast function. We conclude that Brn-3 represents a novel molecular differentiation factor that controls osteoclast maturation and function, suggesting an important role in bone metabolism.


Asunto(s)
Proteínas de Homeodominio/fisiología , Osteoclastos/fisiología , Factor de Transcripción Brn-3A/fisiología , Factor de Transcripción Brn-3B/fisiología , Animales , Secuencia de Bases , Resorción Ósea , Diferenciación Celular , Células Cultivadas , Secuencia de Consenso , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos/química , Osteoclastos/citología , Osteoclastos/metabolismo , Sinaptotagmina I/fisiología , Factor de Transcripción Brn-3A/antagonistas & inhibidores , Factor de Transcripción Brn-3A/metabolismo , Factor de Transcripción Brn-3B/antagonistas & inhibidores , Factor de Transcripción Brn-3B/metabolismo , Activación Transcripcional
10.
Int J Cancer ; 118(4): 869-78, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16152597

RESUMEN

The Brn-3b transcription factor has been shown to be overexpressed in human breast cancer cells and contributes toward cell growth regulation. Using micro-arrays, more than 50 cancer-related genes regulated by Brn-3b in human breast cancer cells have been identified. For example, Brn-3b activates the cell cycle regulator CDK4 that provides a mechanism by which Brn-3b controls the growth of breast cancer cells. Here, we show that Brn-3b regulates plakoglobin (gamma-catenin), a member of the catenin family involved in cell-cell adhesion and signal transduction. Brn-3b expression inversely correlates with plakoglobin expression at both mRNA and protein levels in breast cancer cell lines and human breast biopsies. In contrast, no significant correlation was observed between Brn-3b expression and beta-catenin, or between Brn-3b expression and E-cadherin expression. Brn-3b represses the plakoglobin promoter via a Brn-3 consensus binding site contained within the region -965 to -593 relative to the transcriptional start site. Both repression of the promoter and binding of Brn-3b are lost when this site is mutated. To our knowledge, this is the first time that a Brn-3b POU family transcription factor has been shown to regulate a member of the catenin family, which provides insight into the molecular mechanisms by which Brn-3b expression may favour breast cancer progression and tumor invasion.


Asunto(s)
Neoplasias de la Mama/genética , Factor de Transcripción Brn-3B/fisiología , gamma Catenina/biosíntesis , Sitios de Unión , Neoplasias de la Mama/patología , Adhesión Celular , Desmoplaquinas , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Humanos , Análisis por Micromatrices , Invasividad Neoplásica , Regiones Promotoras Genéticas , Transducción de Señal , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...