Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Neuron ; 97(2): 326-340.e4, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29346753

RESUMEN

At the optic chiasm choice point, ipsilateral retinal ganglion cells (RGCs) are repelled away from the midline by guidance cues, including Ephrin-B2 and Sonic Hedgehog (Shh). Although guidance cues are normally produced by cells residing at the choice point, the mRNA for Shh is not found at the optic chiasm. Here we show that Shh protein is instead produced by contralateral RGCs at the retina, transported anterogradely along the axon, and accumulates at the optic chiasm to repel ipsilateral RGCs. In vitro, contralateral RGC axons, which secrete Shh, repel ipsilateral RGCs in a Boc- and Smo-dependent manner. Finally, knockdown of Shh in the contralateral retina causes a decrease in the proportion of ipsilateral RGCs in a non-cell-autonomous manner. These findings reveal a role for axon-axon interactions in ipsilateral RGC guidance, and they establish that remotely produced cues can act at axon guidance midline choice points.


Asunto(s)
Transporte Axonal/fisiología , Axones/fisiología , Proteínas Hedgehog/metabolismo , Proteínas del Tejido Nervioso/fisiología , Quiasma Óptico/embriología , Células Ganglionares de la Retina/metabolismo , Animales , Inmunoglobulina G/fisiología , Ratones , Quiasma Óptico/metabolismo , Transporte de Proteínas , Receptores de Superficie Celular/fisiología , Receptor Smoothened/fisiología
2.
Cell Cycle ; 15(16): 2102-2107, 2016 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-27229128

RESUMEN

The mechanisms leading to brain tumor formation are poorly understood. Using Ptch1+/- mice as a medulloblastoma model, sequential mutations were found to shape tumor evolution. Initially, medulloblastoma preneoplastic lesions display loss of heterozygosity of the Ptch1 wild-type allele, an event associated with cell senescence in preneoplasia. Subsequently, p53 mutations lead to senescence evasion and progression from preneoplasia to medulloblastoma. These findings are consistent with a model where high levels of Hedgehog signaling caused by the loss of the tumor suppressor Ptch1 lead to oncogene-induced senescence and drive p53 mutations. Thus, cell senescence is an important characteristic of a subset of SHH medulloblastoma and might explain the acquisition of somatic TP53 mutations in human medulloblastoma. This mode of medulloblastoma formation contrasts with the one characterizing Li-Fraumeni patients with medulloblastoma, where TP53 germ-line mutations cause chromothriptic genomic instability and lead to mutations in Hedgehog signaling genes, which drive medulloblastoma growth. Here we discuss in detail these 2 alternative mechanisms leading to medulloblastoma tumorigenesis.


Asunto(s)
Senescencia Celular , Proteínas Hedgehog/metabolismo , Meduloblastoma/patología , Animales , Humanos , Meduloblastoma/genética , Mutación/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
3.
Cell Rep ; 14(12): 2925-37, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26997276

RESUMEN

How brain tumors progress from precancerous lesions to advanced cancers is not well understood. Using Ptch1(+/-) mice to study medulloblastoma progression, we found that Ptch1 loss of heterozygosity (LOH) is an early event that is associated with high levels of cell senescence in preneoplasia. In contrast, advanced tumors have evaded senescence. Remarkably, we discovered that the majority of advanced medulloblastomas display either spontaneous, somatic p53 mutations or Cdkn2a locus inactivation. Consistent with senescence evasion, these p53 mutations are always subsequent to Ptch1 LOH. Introduction of a p53 mutation prevents senescence, accelerates tumor formation, and increases medulloblastoma incidence. Altogether, our results show that evasion of senescence associated with Ptch1 LOH allows progression to advanced tumors.


Asunto(s)
Neoplasias Encefálicas/patología , Senescencia Celular , Meduloblastoma/patología , Receptor Patched-1/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Cerebelo/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Metilación de ADN , Progresión de la Enfermedad , Proteínas Hedgehog/metabolismo , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Pérdida de Heterocigocidad , Meduloblastoma/metabolismo , Meduloblastoma/mortalidad , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Mutación Missense , Receptor Patched-1/genética , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...