Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Oncol ; 50(1): 101-110, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27909720

ABSTRACT

Cancer stem cells (CSCs) in glioma are often responsible for relapse and resistance to therapy. The purpose of the present study was to confirm the self-renewal and migration inhibitory effects of tetrandrine (Tet), which is a compound extracted from the dried root of Stephania tetrandra S. Moore, toward glioma stem-like cells (GSLCs) and to examine the associated molecular mechanisms. Using a neurosphere culture technique, we enriched the GSLC population from the human glioblastoma cell lines U87 and U251. Cells were analyzed using cell counting kit-8 (CCK-8), western blotting, flow cytometry, transwell assay and immunofluorescence staining. GSLCs displayed properties of neural stem cells, including elevated expression of the cancer stem cell marker ALDH1 and ß-catenin. We found that Tet treatment decreased sphere formation in GSLCs in a dose-dependent manner using tumor spheroid formation assay. The GSK3ß inhibitor BIO maintained sphere formation and migration capacity in GSLCs, whereas the ß-catenin/TCF transcription inhibitor ICG-001 decreased sphere formation and the migration capacity of GSLCs. The proportion of apoptotic GSLCs also increased in response to ICG-001 treatment. These results indicate that ß-catenin activity is vital in maintaining neural stem cell traits of GSLCs. Tet inhibits cell viability, neurosphere formation and migration of GSLCs in vitro. Importantly, Tet treatment significantly repressed the nuclear translocation and expression of ß-catenin and induced apoptosis in GSLCs, as indicated in part by the upregulation of Bax, the cleavage of PARP and the downregulation of Bcl-2. The present study demonstrates that the inhibition of ß-catenin in CSCs by Tet could be an effective strategy for the treatment of glioma.


Subject(s)
Benzylisoquinolines/administration & dosage , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , beta Catenin/biosynthesis , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/pathology , Humans , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Pyrimidinones/administration & dosage , bcl-2-Associated X Protein/biosynthesis , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
2.
Eur J Pharmacol ; 764: 228-239, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26086859

ABSTRACT

Tetrandrine (Tet), a bisbenzylisoquinoline alkaloid, has been reported to possess anti-tumour activity. However, its effects on human glioma remain unknown. In this study, we demonstrated that Tet inhibited human glioma cell growth in vitro and in vivo. It has been hypothesised that Tet inhibits glioma growth by affecting glioma cell survival, proliferation and vasculature in and around the xenograft tumour in the chick CAM model and signal transducer and activator of transcription 3 (STAT3) mediated these activities. Therefore, we conducted a detailed analysis of the inhibitory effects of Tet on cell survival using a TUNEL assay and flow cytometric analysis; on cell proliferation based on the expression of proliferating cell nuclear antigen; and on angiogenesis using a CAM anti-angiogenesis assay. We used western blotting to investigate the role of STAT3 on the anti-glioma activities of Tet. The results revealed that Tet inhibited survival and proliferation in human glioma cells, impaired tumour angiogenesis and decreased the expression of phosphorylated STAT3 and its downstream proteins. In sum, our data indicate that STAT3 is involved in Tet-induced the regression of glioma growth by activating tumour cell apoptosis, inhibiting glioma cell proliferation and inhibiting angiogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzylisoquinolines/pharmacology , Brain Neoplasms/metabolism , Glioma/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Benzylisoquinolines/therapeutic use , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chick Embryo , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/physiology , Glioma/drug therapy , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism
3.
J Neurooncol ; 121(1): 41-52, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25173233

ABSTRACT

RATIONAL: Combination therapy to inhibit cancer stem cells may have important clinical implications. Here, we examine the molecular mechanisms by which epigallocatechin gallate (EGCG), a bioactive polyphenol in green tea, inhibits the stem cell characteristics of glioma stem-like cells (GSLCs) and synergizes with temozolomide (TMZ), a DNA-methylating agent commonly used as first-line chemotherapy in gliomas. GSLCs were enriched from the human glioblastoma cell line U87 using neurosphere culture. Cells were analyzed using flow cytometry, quantitative PCR, and western blotting. Compared to U87 cells, a higher percentage of U87 GSLCs remained in the G0/G1 phase, with downregulation of the cell-cycle protein CylinD1 and overexpression of stem cell markers CD133 and ALDH1. The drug-resistance gene ABCB1 (but not ABCG2 or MGMT) also showed high mRNA and protein expression. The resistance index of U87 GSLCs against TMZ and carmustine (BCNU) was 3.0 and 16.8, respectively. These results indicate that U87 GSLCs possess neural stem cell and drug-resistance properties. Interestingly, EGCG treatment inhibited cell viability, neurosphere formation, and migration in this cell model. EGCG also induced apoptosis, downregulation of p-Akt and Bcl-2, and cleaving PARP in a dose-dependent manner. Importantly, EGCG treatment significantly downregulated P-glycoprotein expression but not that of ABCG2 or MGMT and simultaneously enhanced sensitivity to TMZ. Our study demonstrates that the use of EGCG alone or in combination with TMZ may be an effective therapeutic strategy for glioma.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Catechin/analogs & derivatives , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Glioma/physiopathology , Aldehyde Dehydrogenase 1 Family , Animals , Apoptosis/drug effects , Apoptosis/physiology , Catechin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Drug Synergism , Humans , Isoenzymes/metabolism , Neoplastic Stem Cells , RNA, Messenger/metabolism , Rats , Retinal Dehydrogenase/metabolism , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL
...