Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Sci Rep ; 6: 22867, 2016 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-26965927

RESUMEN

We report successful retinal cone enrichment and transplantation using a novel cone-GFP reporter mouse line. Using the putative cone photoreceptor-enriched transcript Coiled-Coil Domain Containing 136 (Ccdc136) GFP-trapped allele, we monitored developmental reporter expression, facilitated the enrichment of cones, and evaluated transplanted GFP-labeled cones in wildtype and retinal degeneration mutant retinas. GFP reporter and endogenous Ccdc136 transcripts exhibit overlapping temporal and spatial expression patterns, both initiated in cone precursors of the embryonic retina and persisting to the adult stage in S and S/M opsin(+) cones as well as rod bipolar cells. The trapped allele does not affect cone function or survival in the adult mutant retina. When comparing the integration of GFP(+) embryonic cones and postnatal Nrl(-/-) 'cods' into retinas of adult wildtype and blind mice, both cell types integrated and exhibited a degree of morphological maturation that was dependent on donor age. These results demonstrate the amenability of the adult retina to cone transplantation using a novel transgenic resource that can advance therapeutic cone transplantation in models of age-related macular degeneration.

3.
J Biol Chem ; 288(6): 4389-404, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23266826

RESUMEN

The inhibition of MyoD expression is important for obtaining muscle progenitors that can replenish the satellite cell niche during muscle repair. Progenitors could be derived from either embryonic stem cells or satellite cells. Hedgehog (Hh) signaling is important for MyoD expression during embryogenesis and adult muscle regeneration. To date, the mechanistic understanding of MyoD regulation by Hh signaling is unclear. Here, we demonstrate that the Hh effector, Gli2, regulates MyoD expression and associates with MyoD gene elements. Gain- and loss-of-function experiments in pluripotent P19 cells show that Gli2 activity is sufficient and required for efficient MyoD expression during skeletal myogenesis. Inhibition of Hh signaling reduces MyoD expression during satellite cell activation in vitro. In addition to regulating MyoD expression, Hh signaling regulates MyoD transcriptional activity, and MyoD activates Hh signaling in myogenic conversion assays. Finally, Gli2, MyoD, and MEF2C form a protein complex, which enhances MyoD activity on skeletal muscle-related promoters. We therefore link Hh signaling to the function and expression of MyoD protein during myogenesis in stem cells.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas Hedgehog/metabolismo , Proteína MioD/biosíntesis , Células Madre Pluripotentes/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Proteínas Hedgehog/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Desarrollo de Músculos/fisiología , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Células Madre Pluripotentes/citología , Células Satélite del Músculo Esquelético/citología , Proteína Gli2 con Dedos de Zinc
4.
J Neurosci ; 31(13): 5169-80, 2011 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-21451052

RESUMEN

The morphogen sonic hedgehog (Shh) plays a crucial role in development of the CNS, including the neural retina. Suppressor of fused (Sufu) has been recently identified as a critical regulator of Hh signaling in mammals. However, the precise roles that Sufu plays in the regulation of proliferation and cell-fate decisions in neural progenitors is unknown. Here, we have addressed these questions by conditionally deleting Sufu in mouse multipotent retinal progenitor cells (RPCs). Sufu deletion in RPCs results in transient increases in Hh activity and proliferation followed by developmentally premature cell-cycle exit. Importantly, we demonstrate a novel role for Sufu in the maintenance of multipotency in RPCs. Sufu-null RPCs downregulate transcription factors required to specify or maintain RPC identity (Rax, Vsx2) and multipotency (Pax6) but continue to express the neural progenitor marker Sox2. These cells fail to express retinal lineage-specific transcription factors, such as Math5, and adopt an amacrine or horizontal cell fate at the expense of all other classes of retinal neurons. Genetic elimination of Gli2 in Sufu-null RPCs attenuates Hh pathway activity and restores multipotency in neural progenitors. These data provide novel evidence that Sufu-mediated antagonism of Hh/Gli2 signaling is required to maintain RPC multipotency and identity.


Asunto(s)
Células Madre Multipotentes/fisiología , Neuronas/fisiología , Proteínas Represoras/fisiología , Retina/embriología , Animales , Diferenciación Celular/genética , Proliferación Celular , Regulación hacia Abajo/genética , Femenino , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Neuronas/citología , Neuronas/metabolismo , Embarazo , Distribución Aleatoria , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Retina/citología , Retina/fisiología , Transducción de Señal/genética
5.
J Cell Biol ; 184(1): 101-12, 2009 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-19124651

RESUMEN

Sonic hedgehog (Shh) is an indispensable, extrinsic cue that regulates progenitor and stem cell behavior in the developing and adult mammalian central nervous system. Here, we investigate the link between the Shh signaling pathway and Hes1, a classical Notch target. We show that Shh-driven stabilization of Hes1 is independent of Notch signaling and requires the Shh effector Gli2. We identify Gli2 as a primary mediator of this response by showing that Gli2 is required for Hh (Hedgehog)-dependent up-regulation of Hes1. We also show using chromatin immunoprecipitation that Gli2 binds to the Hes1 promoter, which suggests that Hes1 is a Hh-dependent direct target of Gli2 signaling. Finally, we show that Shh stimulation of progenitor proliferation and cell diversification requires Gli2 and Hes1 activity. This paper is the first demonstration of the mechanistic and functional link between Shh, Gli, and Hes1 in the regulation of progenitor cell behavior.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Proteínas Hedgehog/fisiología , Proteínas de Homeodominio/fisiología , Receptores Notch/fisiología , Retina/citología , Células Madre/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Proliferación Celular , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Transgénicos , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Receptores Notch/química , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Retina/crecimiento & desarrollo , Transducción de Señal , Factor de Transcripción HES-1 , Proteína Gli2 con Dedos de Zinc
6.
Brain Res ; 1228: 27-42, 2008 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-18625210

RESUMEN

The development of glial precursor cells in the mammalian optic nerve depends on retinal ganglion cell (RGC) axons, but the signals that mediate this neuron-to-glia interaction have not been fully characterized. Sonic hedgehog (Shh) is expressed by RGCs, and we showed previously that it is required for the specification of astrocyte lineage cells at the optic disc. To study the role of RGC-derived Shh on astrocyte development at later developmental stages, we generated mice with a conditional ablation of Shh in the peripheral retina and analyzed gene expression and glial cell development in the optic nerve. Astrocyte development was initiated in the optic nerves of these mutant mice; however, the expression of Hedgehog (Hh) target genes, Gli1 and Ptch1 and cell cycle genes, Ccnd1 and Cdc25b in the optic nerves were downregulated. Astrocyte proliferation was markedly reduced. Oligodendrocyte precursor cells were fewer in the optic nerves of mutant mice, possibly as a consequence of reduced secretion of growth factors by astrocytes. At a later developmental stage, optic nerve axons displayed signs of Wallerian degeneration, including reduction of astrocyte processes, degenerating glial cells and formation of distended axons. We also demonstrate that the Hh pathway can be activated in optic nerve-derived astrocytes in vitro, but fails to induce cell cycle gene expression and proliferation. RGC-derived Shh signalling isthus necessary in vivo for maintenance of astrocyte proliferation, affecting both axo-glial and normal glial cell development in the optic nerve.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Proteínas Hedgehog/fisiología , Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Células Cultivadas , Ciclina D , Ciclinas/genética , Ciclinas/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Inmunohistoquímica , Hibridación in Situ , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Neuroglía/citología , Neuroglía/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Nervio Óptico/crecimiento & desarrollo , Nervio Óptico/ultraestructura , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína con Dedos de Zinc GLI1 , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
7.
Dev Biol ; 308(1): 54-67, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17574231

RESUMEN

The epithelial layers of the ciliary body (CB) and iris are non-neural structures that differentiate from the anterior region of the eyecup, the ciliary margin (CM). We show here that activation of the canonical Wnt signaling pathway is sufficient and necessary for the normal development of anterior eye structures. Pharmacological activation of beta-catenin signaling with lithium (Li(+)) treatment in retinal explants in vitro induced the ectopic expression of the CM markers Otx1 and Msx1. Cre-mediated stabilization of beta-catenin expression in the peripheral retina in vivo induced a cell autonomous upregulation of CM markers at the expense of neural retina (NR) markers and inhibited neurogenesis. Consistent with a cell autonomous conversion to peripheral eye fates, the proliferation index in the region of the retina that expressed stabilized beta-catenin was identical to the wild-type CM and there was an expansion of CB-like structures at later stages. Conversely, Cre-mediated inactivation of beta-catenin reduced CM marker expression as well as the size of the CM and CB/iris. Aberrant CB development in both mouse models was also associated with a reduction in the number of retinal stem cells in vitro. In summary, activation of canonical Wnt signaling is sufficient to promote the development of peripheral eyecup fates at the expense of the NR and is also required for the normal development of anterior eyecup structures.


Asunto(s)
Retina/embriología , Proteínas Wnt/metabolismo , Animales , Secuencia de Bases , Cuerpo Ciliar/embriología , Cuerpo Ciliar/metabolismo , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Técnicas In Vitro , Operón Lac , Ratones , Ratones Transgénicos , Microftalmía/embriología , Microftalmía/genética , Microftalmía/metabolismo , Retina/metabolismo , Transducción de Señal , Proteínas Wnt/genética , beta Catenina/deficiencia , beta Catenina/genética , beta Catenina/metabolismo
8.
Invest Ophthalmol Vis Sci ; 47(11): 5088-97, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17065530

RESUMEN

PURPOSE: The role of the Wnt[b]/beta-catenin-dependent pathway (canonical Wnt pathway) in the context of retinal development and homeostasis is largely unknown. This study was undertaken to characterize activation of the Wnt canonical pathway and its relevance to cell type populations in the developing and adult retina. METHODS: Tissue from TCF/Lef-LacZ (T-cell-specific transcription factor/lymphoid enhancer-binding factor) transgenic mice was used for monitoring the activation of the canonical Wnt pathway. Lithium (Li(+)) treatment was applied to induce ectopic activation of the TCF/Lef-LacZ reporter gene in retinal explants. Gene expression and retinal cell types were examined by in situ hybridization (ISH) or by immunohistochemistry (IHC). RESULTS: On Li(+) treatment, ectopic expression of the TCF/Lef-LacZ reporter gene was rapidly and dramatically induced in retinal explants. The pattern of TCF/Lef-LacZ reporter gene expression was dynamic throughout retinal development and in the adult retina. There was a distinctive expression pattern in each cellular layer, in the developing ciliary margin (CM), and the prospective ciliary epithelium. In the mature retina, the TCF/Lef-LacZ reporter gene was expressed in subsets of retinal ganglion cells (RGCs) and amacrine cells. The expression of the four TCF/Lef transcription factors overlapped with activation of the TCF/Lef-LacZ reporter. CONCLUSIONS: The TCF/Lef-LacZ transgene is a faithful reporter of canonical Wnt signaling in the retina. The pattern of TCF/Lef-LacZ reporter gene activation and of TCF/Lef transcription factor expression suggests that activation of the canonical Wnt pathway is developmental-stage dependent and is spatially modulated. Our findings also imply the involvement of this pathway in the specification and/or generation of ciliary epithelium, cellular differentiation, axon guidance, and connectivity to targets in the central nervous system and in the maintenance or function of specific retinal neurons in the adult.


Asunto(s)
Retina/embriología , Retina/crecimiento & desarrollo , Transducción de Señal/fisiología , Proteínas Wnt/fisiología , Animales , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Técnicas para Inmunoenzimas , Hibridación in Situ , Compuestos de Litio/farmacología , Factor de Unión 1 al Potenciador Linfoide/genética , Masculino , Ratones , Ratones Transgénicos , Factores de Transcripción/genética , Activación Transcripcional , beta Catenina/genética
9.
Dev Cell ; 11(4): 535-46, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17011492

RESUMEN

The transcriptional mechanisms underlying lineage specification and differentiation of embryonic stem (ES) cells remain elusive. Oct-3/4 (POU5f1) is one of the earliest transcription factors expressed in the embryo. Both the pluripotency and the fate of ES cells depend upon a tight control of Oct-3/4 expression. We report that transgene- or TGFbeta-induced increase in Oct-3/4 mRNA and protein levels in undifferentiated ES cells and at early stages of differentiation triggers expression of mesodermal and cardiac specific genes through Smad2/4. cDNA antisense- and siRNA-mediated inhibition of upregulation of Oct-3/4 in ES cells prevent their specification toward the mesoderm and their differentiation into cardiomyocytes. Similarly, Oct-3/4 siRNA injected in the inner cell mass of blastocysts impairs cardiogenesis in early embryos. Thus, quantitative Oct-3/4 expression is regulated by a morphogen, pointing to a pivotal and physiological function of the POU factor in mesodermal and cardiac commitments of ES cells and of the epiblast.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre/citología , Células Madre/fisiología , Animales , Blastocisto/citología , Blastocisto/metabolismo , Linaje de la Célula , Inmunohistoquímica , Hibridación in Situ , Ratones , Microinyecciones , Miocitos Cardíacos/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Técnicas de Cultivo de Órganos , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/metabolismo
10.
Mol Cell Neurosci ; 32(3): 274-82, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16815712

RESUMEN

The morphogen Sonic hedgehog (Shh) is expressed by the projection neurons of the retina, retinal ganglion cells (RGCs) and promotes retinal precursor cell (RPC) proliferation. To distinguish between direct and indirect effects of Hedgehog (Hh) pathway activation in the perinatal mouse retina, we followed the fate of cells that expressed a constitutively active allele of Smoothened (SMO-M2), the signal transduction component of the Hh pathway. SMO-M2 expression promoted a cell-autonomous increase in CyclinD1 expression and RPC proliferation and promoted the development of cells with an inner nuclear layer identity. SMO-M2 expression also inhibited rhodopsin expression in uninfected cells, thus highlighting an unexpected non-cell autonomous effect of Hh pathway activation on photoreceptor development.


Asunto(s)
Retina/metabolismo , Transducción de Señal/fisiología , Transactivadores/metabolismo , Animales , Células Cultivadas , Proteínas Hedgehog , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Células 3T3 NIH , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Retina/crecimiento & desarrollo , Retina/fisiología , Receptor Smoothened , Transactivadores/genética , Transactivadores/fisiología
11.
Development ; 132(22): 5103-13, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16236765

RESUMEN

The timing of cell cycle exit and temporal changes in the developmental competence of precursor cells are key components for the establishment of the normal complement of cell types in the mammalian retina. The identity of cell extrinsic cues that control these processes is largely unknown. We showed previously in mouse retina that sonic hedgehog (Shh) signalling from retinal ganglion cells (RGCs) to retinal precursor cells (RPC) is required for the establishment of normal retinal organization. Here, we show that conditional ablation of Shh expression in the peripheral mouse results in a depletion of the RPC pool, owing to precocious cell-cycle exit and neuronal differentiation. These changes were correlated with the downregulation of cyclin D1 and Hes1 gene expression. Shh inactivation also results in an increase in RGC number owing to a bias of RPC towards RGC production. In contrast to zebrafish, where Shh signalling drives cell cycle exit and RGC development, our findings indicate that in the mouse retina Shh signalling is required to maintain RPC proliferation and to control the timing of RGC development.


Asunto(s)
Proliferación Celular , Retina/embriología , Células Ganglionares de la Retina/fisiología , Transactivadores/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Regulación hacia Abajo , Proteínas Hedgehog , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Integrasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Ganglionares de la Retina/citología , Transducción de Señal/fisiología , Factor de Transcripción HES-1
12.
J Virol ; 77(13): 7193-201, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12805418

RESUMEN

ts1 is a temperature-sensitive mutant of Moloney murine leukemia virus that induces a rapid spongiform encephalopathy in mice infected as newborns. The pathological features include the formation of ubiquitinated inclusions resembling Lewy bodies. To determine how perturbation of the ubiquitin-proteasome pathway might affect ts1-mediated neurodegeneration, the virus was introduced into transgenic mice in which the assembly of ubiquitin chains was compromised by the expression of dominant-negative mutant ubiquitin. The onset of symptoms was greatly delayed in a transgenic mouse line expressing K48R mutant ubiquitin; no such delay was observed in mice expressing a wild-type ubiquitin transgene or K63R mutant ubiquitin. The extended latency was found to correlate with a delayed increase in viral titers. Pathological findings in K48R transgenic mice at 60 days were found to be similar to those in the other strains at 30 days, suggesting that while delayed, the neurodegenerative process in K48R mice was otherwise similar. These data demonstrate the sensitivity of retroviral replication to the partial disruption of ubiquitin-mediated proteolysis in vivo, a finding that may have therapeutic potential.


Asunto(s)
Encéfalo/patología , Virus de la Leucemia Murina de Moloney/patogenicidad , Mutación , Ubiquitina/fisiología , Animales , Ratones , Ratones Transgénicos , Ubiquitina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...