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.
Int J Oncol ; 52(2): 485-495, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29345288

ABSTRACT

Glioblastoma (GBM) is the most common type of malignant brain tumor. In spite of recent advancements in surgical techniques, chemotherapy, and radiation therapy, patients with GBM often face a dire prognosis. MicroRNAs have been shown to modulate the aggressiveness of various cancers, and have emerged as possible therapeutic agents for the management of GBM. miR­205 is dysregulated in glioma and act as a prognostic indicator. However, the role of miR­205 in the development of GBM has not been elucidated. To better understand the pathogenesis of GBM, we examine the biological significance and molecular mechanisms of miR­205 in GBM cells. Zinc finger E-box binding homeobox 1 (ZEB1) has been shown to regulate the epithelial-mesenchymal transition (EMT), which is strongly associated with GBM malignancy. In the present study, we show miR­205 expression is reduced in GBM tissues and cell lines, and ZEB1 expression is inversely correlated with miR­205 expression. We also show ZEB1 is a downstream target of miR­205 and the Akt/mTOR signaling pathway is activated when miR­205 interacts with ZEB1. Increased activity of miR­205 in GBM cells significantly inhibits migration and invasion, and prevents EMT. Furthermore, overexpression of ZEB1 partially abolishes these inhibitory effects of miR­205. We show that miR­205 negatively regulates the expression of ZEB1 in GBM, inhibits cell migration and invasion, and prevents EMT, at least in part through the inhibition of the activation of the Akt/mTOR signaling pathway. Our results indicate miR­205 may be an efficacious therapeutic agent in the treatment of GBM.


Subject(s)
Brain Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Glioblastoma/pathology , MicroRNAs/genetics , 3' Untranslated Regions , Adult , Aged , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
2.
Oncotarget ; 8(14): 22783-22799, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28187000

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant brain tumor with limited therapeutic options. Temozolomide (TMZ) is a novel cytotoxic agent used as first-line chemotherapy for GBM, however, some individual cells can't be isolated for surgical resection and show treatment-resistance, thus inducing poor prognosis. By using the HiSeq sequencing and bioinformatics methods, we identified lncRNAs showing different expression levels in TMZ-resistant and non-resistant patients. RT-qPCR was then performed in tissues and serum samples, and lncRNA MALAT1 was finally identified to show considerable discriminating potential to identify responding patients from non-responding patients. Moreover, high serum MALAT1 expression was associated with poor chemoresponse and survival in GBM patients receiving TMZ treatment. Subsequently, the TMZ resistant cell lines were established, and the CCK8 assay showed that lncRNA MALAT1 knockdown significantly reversed TMZ resistance in GBM cells. The gain and loss-function experiments revealed that miR-203 was down-regulated by MALAT1 and this interaction has reciprocal effects. Besides, thymidylate synthase (TS) mRNA was identified as a direct target of miR-203. LncRNA MALAT1 inhibition re-sensitized TMZ resistant cells through up-regulating miR-203 and down-regulating TS expression. On the other hand, MALAT1 overexpression promoted resistance by suppressing miR-203 and promoting TS expression. In conclusion, our integrated approach demonstrates that enhanced expression of lncRNA MALAT1 confers a potent poor therapeutic efficacy and inhibition of MALAT1 levels could be a future direction to develop a novel therapeutic strategy to overcome TMZ resistance in GBM patients.


Subject(s)
Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Glioblastoma/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Thymidylate Synthase/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Neoplasm Staging , Prognosis , Survival Rate , Temozolomide , Thymidylate Synthase/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...