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

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Glia ; 66(5): 999-1015, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29392777

RESUMO

Malignant glioma is one of the deadliest types of cancer. Understanding how the cell of origin progressively evolves toward malignancy in greater detail could provide mechanistic insights and lead to novel concepts for tumor prevention and therapy. Previously we have identified oligodendrocyte precursor cell (OPC) as the cell of origin for glioma following the concurrent deletion of p53 and NF1 using a mouse genetic mosaic system that can reveal mutant cells prior to malignancy. In the current study, we set out to deconstruct the gliomagenic process in two aspects. First, we determined how the individual loss of p53 or NF1 contributes to aberrant behaviors of OPCs. Second, we determined how signaling aberrations in OPCs progressively change from pre-malignant to transformed stages. We found that while the deletion of NF1 leads to mutant OPC expansion through increased proliferation and decreased differentiation, the deletion of p53 impairs OPC senescence. Signaling analysis showed that, while PI3K and MEK pathways go through stepwise over-activation, mTOR signaling remains at the basal level in pre-transforming mutant OPCs but is abruptly up-regulated in tumor OPCs. Finally, inhibiting mTOR via pharmacological or genetic methods, led to a significant blockade of gliomagenesis but had little impact on pre-transforming mutant OPCs, suggesting that mTOR is necessary for final transformation but not early progression. In summary, our findings show that deconstructing the tumorigenic process reveals specific aberrations caused by individual gene mutations and altered signaling events at precise timing during tumor progression, which may shed light on tumor-prevention strategies.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Neurofibromina 1/deficiência , Proteína Supressora de Tumor p53/deficiência , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Senescência Celular/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Glioma/patologia , Células HEK293 , Humanos , Camundongos Transgênicos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurofibromina 1/genética , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
2.
Proc Natl Acad Sci U S A ; 111(40): E4214-23, 2014 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-25246577

RESUMO

How malignant gliomas arise in a mature brain remains a mystery, hindering the development of preventive and therapeutic interventions. We previously showed that oligodendrocyte precursor cells (OPCs) can be transformed into glioma when mutations are introduced perinatally. However, adult OPCs rarely proliferate compared with their perinatal counterparts. Whether these relatively quiescent cells have the potential to transform is unknown, which is a critical question considering the late onset of human glioma. Additionally, the premalignant events taking place between initial mutation and a fully developed tumor mass are particularly poorly understood in glioma. Here we used a temporally controllable Cre transgene to delete p53 and NF1 specifically in adult OPCs and demonstrated that these cells consistently give rise to malignant gliomas. To investigate the transforming process of quiescent adult OPCs, we then tracked these cells throughout the premalignant phase, which revealed a dynamic multistep transformation, starting with rapid but transient hyperproliferative reactivation, followed by a long period of dormancy, and then final malignant transformation. Using pharmacological approaches, we discovered that mammalian target of rapamycin signaling is critical for both the initial OPC reactivation step and late-stage tumor cell proliferation and thus might be a potential target for both glioma prevention and treatment. In summary, our results firmly establish the transforming potential of adult OPCs and reveal an actionable multiphasic reactivation process that turns slowly dividing OPCs into malignant gliomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Transformação Celular Neoplásica/metabolismo , Glioma/metabolismo , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Animais , Antineoplásicos/farmacologia , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Perfilação da Expressão Gênica , Glioma/genética , Glioma/patologia , Imuno-Histoquímica , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Oligodendroglia/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Tamoxifeno/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Curr Pathobiol Rep ; 1(1): 19-28, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23538742

RESUMO

Inflammation has been closely linked to various forms of cancer. Less is known about the role of inflammation in glioma, especially at the initiation stage. In this review, we first describe the unique features of the immune system in the brain. We then discuss the current understanding of the mechanisms by which glioma cells modulate the immune system, especially how bi-directional communications between immune cells and glioma cells create an immunosuppressed microenvironment that promotes tumor survival and growth. We also address the potential tumor-initiating roles of inflammation in glioma. Finally, we describe several immunotherapy approaches currently being developed to reverse these interactions and stimulate the immune system to eliminate glioma cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA