Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 12(4): e0175147, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28380052

RESUMO

It has been proposed that a subpopulation of tumour cells with stem cell-like characteristics, known as cancer stem cells (CSCs), drives tumour initiation and generates tumour heterogeneity, thus leading to cancer metastasis, recurrence, and drug resistance. Although there has been substantial progress in CSC research into many solid tumour types, an understanding of the biology of CSCs in lung cancer remains elusive, mainly because of their heterogeneous origins and high plasticity. Here, we demonstrate that engineered lung cancer cells derived from normal human airway basal epithelial cells possessed CSC-like characteristics in terms of multilineage differentiation potential and strong tumour-initiating ability. Moreover, we established an in vitro 3D culture system that allowed the in vivo differentiation process of the CSC-like cells to be recapitulated. This engineered CSC model provides valuable opportunities for studying the biology of CSCs and for exploring and evaluating novel therapeutic approaches and targets in lung CSCs.


Assuntos
Engenharia Celular/métodos , Pulmão/citologia , Células-Tronco Neoplásicas/fisiologia , Mucosa Respiratória/citologia , Animais , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Feminino , Humanos , Pulmão/patologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias
2.
Cancer Res ; 71(7): 2541-9, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21447735

RESUMO

It has been challenging to engineer lung adenocarcinoma models via oncogene-mediated transformation of primary cultured normal human cells. Although viral oncoprotein-mediated malignant transformation has been reported, xenografts derived from such transformed cells generally represent poorly differentiated cancers. Here, we demonstrate that the combined expression of multiple cellular factors induces malignant transformation in normal human lung epithelial cells. Although a combination of four genetic alterations, including hTERT overexpression, inactivation of the pRB and p53 pathways, and KRAS activation, is insufficient for normal human small airway epithelial cells to be fully transformed, expression of one additional oncogene induces malignant transformation. Notably, we have succeeded in reproducing human lung adenocarcinoma phenotypes in the flanks of nude mice by introducing an active form of PIK3CA, CYCLIN-D1, or a dominant-negative form of LKB1 in combination with the four genetic alterations above. Besides differentiated lung cancer, poorly differentiated cancer models can also be engineered by employing c-MYC as one of the genetic elements, indicating that histologic features and degree of differentiation of xenografts are controllable to some extent by changing the combination of genetic elements introduced. This is the first study reporting malignant transformation of normal lung epithelial cells in the absence of viral oncoproteins. We propose that our model system would be useful to identify the minimal and most crucial set of changes required for lung tumorigenesis, and that it would provide a broadly applicable approach for discovering attractive therapeutic targets.


Assuntos
Transformação Celular Viral/fisiologia , Retroviridae/fisiologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma/virologia , Adenocarcinoma de Pulmão , Adulto , Animais , Diferenciação Celular/fisiologia , Transformação Celular Viral/genética , Quinase 4 Dependente de Ciclina/biossíntese , Quinase 4 Dependente de Ciclina/genética , Transição Epitelial-Mesenquimal , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/virologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fenótipo , Retroviridae/genética , Telomerase/biossíntese , Telomerase/genética , Transplante Heterólogo , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Adulto Jovem
3.
Mol Cell Biol ; 30(19): 4604-15, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20679479

RESUMO

The small GTPase Rho regulates cell morphogenesis through remodeling of the actin cytoskeleton. While Rho is overexpressed in many clinical cancers, the role of Rho signaling in oncogenesis remains unknown. mDia1 is a Rho effector producing straight actin filaments. Here we transduced mouse embryonic fibroblasts from mDia1-deficient mice with temperature-sensitive v-Src and examined the involvement and mechanism of the Rho-mDia1 pathway in Src-induced oncogenesis. We showed that in v-Src-transduced mDia1-deficient cells, formation of actin filaments is suppressed, and v-Src in the perinuclear region does not move to focal adhesions upon a temperature shift. Consequently, membrane translocation of v-Src, v-Src-induced morphological transformation, and podosome formation are all suppressed in mDia1-deficient cells with impaired tyrosine phosphorylation. mDia1-deficient cells show reduced transformation in vitro as examined by focus formation and colony formation in soft agar and exhibit suppressed tumorigenesis and invasion when implanted in nude mice in vivo. Given overexpression of c-Src in various cancers, these findings suggest that Rho-mDia1 signaling facilitates malignant transformation and invasion by manipulating the actin cytoskeleton and targeting Src to the cell periphery.


Assuntos
Proteínas de Transporte/metabolismo , Transformação Celular Neoplásica , Neoplasias Experimentais/metabolismo , Proteína Oncogênica pp60(v-src)/metabolismo , Animais , Proteínas de Transporte/genética , Proliferação de Células , Células Cultivadas , Citoesqueleto/metabolismo , Embrião de Mamíferos/citologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Forminas , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Immunoblotting , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Microscopia de Fluorescência , Microscopia de Vídeo , Invasividade Neoplásica , Transplante de Neoplasias , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Proteína Oncogênica pp60(v-src)/genética , Transfecção
4.
J Cell Biol ; 189(5): 901-17, 2010 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-20513769

RESUMO

LL5beta has been identified as a microtubule-anchoring factor that attaches EB1/CLIP-associating protein (CLASP)-bound microtubule plus ends to the cell cortex. In this study, we show that LL5beta and its homologue LL5alpha (LL5s) colocalize with autocrine laminin-5 and its receptors, integrins alpha3beta1 and alpha6beta4, at the basal side of fully polarized epithelial sheets. Depletion of both laminin receptor integrins abolishes the cortical localization of LL5s, whereas LL5 depletion reduces the amount of integrin alpha3 at the basal cell cortex. Activation of integrin alpha3 is sufficient to initiate LL5 accumulation at the cell cortex. LL5s form a complex with the cytoplasmic tails of these integrins, but their interaction might be indirect. Analysis of the three-dimensional distribution of microtubule growth by visualizing EB1-GFP in epithelial sheets in combination with RNA interference reveals that LL5s are required to maintain the density of growing microtubules selectively at the basal cortex. These findings reveal that signaling from laminin-integrin associations attaches microtubule plus ends to the epithelial basal cell cortex.


Assuntos
Proteínas de Transporte/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Laminina/metabolismo , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Proteínas de Transporte/genética , Adesão Celular , Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Feminino , Humanos , Integrina alfa3/genética , Integrina alfa3/metabolismo , Integrina alfa3beta1/genética , Integrina alfa3beta1/metabolismo , Integrina alfa6/genética , Integrina alfa6/metabolismo , Integrina alfa6beta4/genética , Integrina alfa6beta4/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Laminina/genética , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/química , Proteínas do Tecido Nervoso/genética , Ligação Proteica/fisiologia , RNA Interferente Pequeno/genética , Receptores de Laminina/genética , Receptores de Laminina/metabolismo
5.
Proc Natl Acad Sci U S A ; 103(14): 5490-5, 2006 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-16569692

RESUMO

Human diploid fibroblasts (HDF) immortalized by hTERT and simian virus 40 (SV40) early region (ER) exhibit a limited degree of transformation upon the expression of activated H-RAS (H-RAS V12) compared with rat embryonic fibroblasts (REF) immortalized by SV40 ER. Here, we identified FRA1 as a determinant for this difference in RAS-induced transformation. FRA1 was not induced by H-RAS V12 in the immortalized HDF, in contrast to its marked accumulation in the immortalized REF. Ectopic expression of FRA1 significantly enhanced anchorage-independent growth of various HDF expressing hTERT, SV40 ER, and H-RAS V12. More importantly, FRA1 could induce anchorage-independent growth as well as nude mice tumor formation of the immortalized HDF in the absence of H-RAS V12. The results of an in vitro kinase assay clearly showed that the RAS-induced extracellular signal-regulated kinase (ERK) activation, which is responsible for FRA1 induction, was markedly attenuated in the HDF compared with that in the REF, despite no obvious differences in the phosphorylation status of ERK between the species. Our results strongly suggest that HDF negatively regulate the mitogen-activated protein kinase kinase (MEK)/ERK pathway more efficiently than REF, and consequently express less malignant phenotypes in response to H-RAS V12.


Assuntos
Genes ras , Proteínas Proto-Oncogênicas c-fos/fisiologia , Animais , Western Blotting , Transformação Celular Neoplásica , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ratos
6.
Oncogene ; 24(36): 5648-55, 2005 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-16007212

RESUMO

Ras-induced transformation is characterized not only by uncontrolled proliferation but also by drastic morphological changes accompanied by the disruption of the actin cytoskeleton. Previously, we reported that human fibroblasts are more resistant than rodent fibroblasts to Ras-induced transformation. To explore the molecular basis for the difference in susceptibility to Ras-induced transformation, we investigated the effect of activated H-Ras on the actin cytoskeleton in human diploid fibroblasts and in rat embryo fibroblasts, both of which are immortalized by SV40 early region. We demonstrate here that Ras-induced morphological changes, decreased expression of tropomyosin isoforms, and suppression of the ROCK/LIMK/Cofilin pathway observed in the rat fibroblasts were not detected in the human fibroblasts even with high expression levels of Ras. We also show that activation of the MEK/ERK pathway sufficed to induce all of these alterations in the rat fibroblasts, whereas the human fibroblasts were refractory to these MEK/ERK-mediated changes. In addition to morphological changes, we demonstrated that the expression of activated Ras induced an invasive phenotype in the rat, but not in the human fibroblasts. These studies provide evidence for the existence of human-specific mechanisms that resist Ras/MEK/ERK-mediated transformation.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Diploide , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica/patologia , Proteínas ras/metabolismo , Fatores de Despolimerização de Actina , Animais , Células Cultivadas , Regulação para Baixo , Matriz Extracelular/metabolismo , Fibroblastos , Humanos , Sistema de Sinalização das MAP Quinases , Proteínas dos Microfilamentos/metabolismo , Peso Molecular , Fosforilação , Isoformas de Proteínas/metabolismo , Ratos , Tropomiosina/metabolismo , Proteínas ras/genética
7.
Oncogene ; 23(52): 8527-34, 2004 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-15378021

RESUMO

The c-abl tyrosine kinase is the proto-oncogene of the v-abl oncogene of the Abelson murine leukemia virus. Although mutational variants of c-Abl can exhibit gain of function and can produce a transformed phenotype, the function of c-Abl in transformation remained unclear. Here, we report that the loss of c-abl facilitates transformation. c-abl-knockout mouse embryonic fibroblasts (MEFs) immortalized by SV40 T antigen acquired anchorage-independent growth, and by constructing mutational variants of T antigen we showed that binding of large T antigen to p53 and RB was necessary to induce anchorage-independent growth. Although c-abl/p53 double-knockout MEFs did not undergo anchorage-independent growth, those expressing human papilloma virus 16 E7, which mainly inactivates RB, did. Our results show that the loss of c-abl facilitates anchorage-independent growth in the context of p53 and RB deficiency, and suggest that loss of function of c-abl facilitates some types of transformation.


Assuntos
Fibroblastos/metabolismo , Proteínas Proto-Oncogênicas c-abl/deficiência , Proteína do Retinoblastoma/deficiência , Proteína Supressora de Tumor p53/deficiência , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Camundongos , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo
8.
J Biol Chem ; 278(24): 21685-92, 2003 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12672821

RESUMO

Mammalian Enabled (Mena) is a mammalian homologue of Drosophila Enabled (Ena), which genetically interacts with Drosophila Abl tyrosine kinase. The signaling pathway involving c-Abl and Mena (Ena) is not fully understood. To find molecules that participate in the c-Abl/Mena pathway, we searched for Mena-binding proteins using a yeast two-hybrid system. We identified Abl interactor 1 (Abi-1), which is known to interact with c-Abl, as a binding protein for Mena. Binding analysis revealed that the Ena/Vasp homology 1 domain of Mena and the polyproline structure of Abi-1 are necessary for the interaction. The interaction between Mena and Abi-1 was also observed in a mammalian expression system. Importantly, Abi-1 dramatically promoted c-Abl-mediated tyrosine phosphorylation of Mena but not other substrates such as c-Cbl. Mutational analysis demonstrated that the phosphorylation site of Mena is Tyr-296. Our results suggest that Abi-1 regulates c-Abl-mediated phosphorylation of Mena by interacting with both proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/fisiologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Tirosina/química , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte/fisiologia , Linhagem Celular , Análise Mutacional de DNA , DNA Complementar/metabolismo , Biblioteca Gênica , Glutationa Transferase/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Proteínas dos Microfilamentos , Microscopia de Fluorescência , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...