Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Biol ; 22(8): 2703-15, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11909963

ABSTRACT

Scatter factor/hepatocyte growth factor (SF/HGF) expression has been linked to malignant progression in glial neoplasms. Using two glioma cell lines, U373MG and SNB-19, we have demonstrated that SF/HGF stimulation allows cells to escape G(1)/G(0) arrest induced by contact inhibition or serum withdrawal. SF/HGF induced effects on two mechanisms of cell cycle regulation: suppression of the cyclin-dependent kinase inhibitor p27 and induction of the transcription factor c-Myc. Regulation of p27 by SF/HGF was posttranslational and is associated with p27 nuclear export. Transient transfections of U373MG and SNB-19 with wild-type p27 and a degradation-resistant p27T187A mutant were insufficient to induce cell cycle arrest, and SF/HGF downregulation of p27 was not necessary for cell cycle reentry. Analysis of Cdk2 kinase activity and p27 binding to cyclin E complexes in the presence of exogenous wild-type p27 or p27T187A demonstrated that Cdk2 activity was not necessary for SF/HGF-mediated G(1)/S transition. Similarly, overexpression of dominant-negative forms of Cdk2 did not block SF/HGF-triggered cell cycle progression. In contrast, SF/HGF transcriptionally upregulated c-Myc, and overexpression of c-Myc was able to prevent G(1)/G(0) arrest in the absence of SF/HGF. Transient overexpression of MadMyc, a dominant-negative chimera for c-Myc, caused G(1)/G(0) arrest in logarithmically growing cells and blocked SF/HGF-mediated G(1)/S transition. c-Myc did not exert its effects through p27 downregulation in these cell lines. SF/HGF induced E2F1-dependent transcription, the inhibition of which did not block SF/HGF-induced cell cycle progression. We conclude that SF/HGF prevents G(1)/G(0) arrest in glioma cell lines by a c-myc-dependent mechanism that is independent of p27, Cdk2, or E2F1.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/drug effects , DNA-Binding Proteins , Glioblastoma/metabolism , Glioblastoma/pathology , Hepatocyte Growth Factor/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Active Transport, Cell Nucleus , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Contact Inhibition , Culture Media , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , G1 Phase/drug effects , G1 Phase/physiology , Glioblastoma/genetics , Hepatocyte Growth Factor/physiology , Humans , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , S Phase/drug effects , S Phase/physiology , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
2.
FASEB J ; 16(1): 108-10, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11729097

ABSTRACT

The multifunctional growth factor scatter factor/hepatocyte growth factor (SF/HGF) and its receptor c-met have been implicated in the genesis, malignant progression, and chemo/radioresistance of multiple human malignancies, including gliomas. We examined the antitumor effects of targeting SF/HGF and c-met expression in pre-established glioma xenografts by using novel chimeric U1snRNA/ribozymes. Transient expression of anti-SF/HGF and anti-c-met U1snRNA/ribozymes inhibited SF/HGF and c-met expression, c-met receptor activation, tumor cell migration, and anchorage-independent colony formation in vitro. Delivery of U1snRNA/ribozymes to established subcutaneous glioma xenografts via liposome-DNA complexes significantly inhibited tumor growth as well as tumor SF/HGF and c-met expression levels. Histologic analysis of tumors treated with U1snRNA/ribozymes showed a significant decrease in blood vessel density, an increase in activation of the pro-apoptotic enzyme caspase-3, and an increase in tumor cell apoptosis. Treatment of animals bearing intracranial glioma xenografts with anti-SF/HGF and anti-c-met U1snRNA/ribozymes by either intratumoral injections of adenoviruses expressing the transgenes or intravenous injections of U1snRNA/ribozyme-liposome complexes substantially inhibited tumor growth and promoted animal survival. We demonstrate that SF/HGF and/or c-met expression can be targeted in vivo to inhibit tumor growth. In addition, our findings represent the first in vivo application of chimeric U1snRNA/ribozymes, which have numerous potential therapeutic gene-targeting applications.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Hepatocyte Growth Factor/genetics , Proto-Oncogene Proteins c-met/genetics , RNA, Catalytic/genetics , RNA, Small Nuclear/genetics , Adenoviridae/genetics , Animals , Apoptosis , Base Sequence , Brain Neoplasms/pathology , Cell Division , DNA, Antisense/administration & dosage , Genetic Vectors , Glioblastoma/pathology , Hepatocyte Growth Factor/biosynthesis , Humans , Mice , Molecular Sequence Data , Neovascularization, Pathologic , Proto-Oncogene Proteins c-met/metabolism , RNA, Neoplasm/analysis , Transgenes , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...