Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Cycle ; 15(16): 2102-2107, 2016 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-27229128

RESUMO

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.


Assuntos
Senescência Celular , Proteínas Hedgehog/metabolismo , Meduloblastoma/patologia , Animais , Humanos , Meduloblastoma/genética , Mutação/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
2.
Cell Rep ; 14(12): 2925-37, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26997276

RESUMO

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.


Assuntos
Neoplasias Encefálicas/patologia , Senescência Celular , Meduloblastoma/patologia , Receptor Patched-1/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Cerebelo/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Metilação de DNA , Progressão da Doença , Proteínas Hedgehog/metabolismo , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Perda de Heterozigosidade , Meduloblastoma/metabolismo , Meduloblastoma/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Mutação de Sentido Incorreto , Receptor Patched-1/genética , Regiões Promotoras Genéticas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA