Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Asian Pac J Cancer Prev ; 20(1): 23-32, 2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30677865

ABSTRACT

The Protein kinase CK2 (formerly known as casein kinase 2) is a highly conserved serine/ threonine kinase overexpressed in various human carcinomas and its high expression often correlates with poor prognosis. CK2 protein is localized in the nucleus of many tumor cells and correlates with clinical features in many cases. Increased expression of CK2 in mice results in the development of various types of carcinomas (both solids and blood related tumors, such as (breast carcinoma, lymphoma, etc), which reveals its carcinogenic properties. CK2 plays essential roles in many key biological processes related to carcinoma, including cell apoptosis, DNA damage responses and cell cycle regulation. CK2 has become a potential anti-carcinoma target. Various CK2 inhibitors have been developed with anti-neoplastic properties against a variety of carcinomas. Some CK2 inhibitors have showed good results in in vitro and pre-clinical models, and have even entered in clinical trials. This article will review effects of CK2 and its inhibitors on common carcinomas in in vitro and pre-clinical studies.


Subject(s)
Antineoplastic Agents/therapeutic use , Casein Kinase II/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Animals , Disease Management , Humans , Neoplasms/pathology , Prognosis
4.
Cancer Lett ; 379(1): 134-42, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27267806

ABSTRACT

Histone deacetylases are considered to be among the most promising targets in drug development for cancer therapy. Histone deacetylase 6 (HDAC6) is a unique cytoplasmic enzyme that regulates many biological processes involved in tumorigenesis through its deacetylase and ubiquitin-binding activities. Here, we report that HDAC6 is overexpressed in glioblastoma tissues and cell lines. Overexpression of HDAC6 promotes the proliferation and spheroid formation of glioblastoma cells. HDAC6 overexpression confers resistance to temozolomide (TMZ) mediated cell proliferation inhibition and apoptosis induction. Conversely, knockdown of HDAC6 inhibits cell proliferation, impairs spheroid formation and sensitizes glioblastoma cells to TMZ. The inhibition of HDAC6 deacetylase activity by selective inhibitors inhibits the proliferation of glioblastoma cells and induces apoptosis. HDAC6 selective inhibitors can sensitize glioblastoma cells to TMZ. Moreover, we showed that HDAC6 mediated EGFR stabilization might partly account for its oncogenic role in glioblastoma. TMZ resistant glioblastoma cells showed higher expression of HDAC6 and more activation of EGFR. HDAC6 inhibitors decrease EGFR protein levels and impair the activation of the EGFR pathway. Taken together, our results suggest that the inhibition of HDAC6 may be a promising strategy for the treatment of glioblastoma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Histone Deacetylases/metabolism , Apoptosis/drug effects , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Protein Stability , Signal Transduction/drug effects , Temozolomide , Time Factors , Transfection , Up-Regulation
5.
Biochem Cell Biol ; 94(3): 247-55, 2016 06.
Article in English | MEDLINE | ID: mdl-27105323

ABSTRACT

Transcription factor 3 (TCF3) is a member of the T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factor family. Recent studies have demonstrated its potential carcinogenic properties. Here we show that TCF3 was upregulated in glioma tissues compared with normal brain tissues. This upregulation of the TCF3 gene probably has functional significance in brain-tumor progression. Our studies on glioblastoma multiforme (GBM) cell lines show that knock-down of TCF3 induced apoptosis and inhibited cell migration. Further analysis revealed that down-regulation of TCF3 gene expression inhibits Akt and Erk1/2 activation, suggesting that the carcinogenic properties of TCF3 in GBM are partially mediated by the phosphatidylinositol 3-kinase-Akt and MAPK-Erk signaling pathways. Considered together, the results of this study demonstrate that high levels of TCF3 in gliomas potentially promote glioma development through the Akt and Erk pathways.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/pathology , Glioma/pathology , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation
6.
Oncotarget ; 7(8): 9222-35, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26824182

ABSTRACT

Abnormalities of autophagy have been implicated in an increasing number of human cancers, including glioma. To date, there is a wealth of evidence indicating that microRNAs (miRNAs) contribute significantly to autophagy in a variety of cancers. Previous studies have suggested that miR-129 functioned as an important inhibitor of the cell cycle and could promote the apoptosis of many cancer cell lines in vitro. Here, we reported that miR-129 acted as a potent inducer of autophagy. Forced expression of miR-129 could induce autophagic flux by targetedly suppressing Notch-1 in glioma cells. The autophagy induced by miR-129 could restrain the activity of mammalian target of rapamycin (mTOR) and upregulate Beclin-1. Moreover, we demonstrated that E2F transcription factor 7 (E2F7) could also trigger autophagic flux by upregulating Beclin-1 and mediating miR-129-induced autophagy. Additionally, knockdown of Notch-1 could upregulate the expression of E2F7, whereas downregulation of E2F7 alleviated shNotch-1-induced autophagic flux. In particular, knockdown of endogenous Beclin-1 could effectively reduce autophagic flux stimulated by miR-129 and E2F7. Interestingly, upon attenuation of miR-129- or E2F7-triggered autophagic flux rescued cell viability suppressed by them. More importantly, intratumoral injection of pHAGE-miR-129 lentivirus in a nude mouse xenograft model significantly restrained tumor growth and triggered autophagy. In conclusion, these findings identify a new function for miR-129 as a potent inducer of autophagy through a novel Notch-1/E2F7/Beclin-1 axis in glioma.


Subject(s)
Autophagy/genetics , Beclin-1/metabolism , E2F7 Transcription Factor/metabolism , Glioma/genetics , MicroRNAs/genetics , Receptor, Notch1/metabolism , Animals , Beclin-1/genetics , Cell Line , Glioma/pathology , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transplantation, Heterologous , Up-Regulation
7.
Int J Oncol ; 47(4): 1603-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26323558

ABSTRACT

Treatment with cisplatin, a chemotherapeutic agent commonly used in glioma patients, often results in chemoresistance. Increasing evidence has shown that microRNAs (miRNAs) are implicated in the drug resistance of gliomas. However, the function of miR­873 in cisplatin resistance of gliomas remains unknown. In this study, we found that many miRNAs, including miR­873, are differentially expressed in cisplatin-resistant glioma cells compared to wild-type glioma cells. Moreover, cisplatin reduced the expression of miR­873 in a time-dependent manner. Overexpression of miR­873 decreased the cell proliferation, migration and invasion while increased apoptosis of cisplatin-resistant glioma cells and sensitized the cells to cisplatin-induced cell growth arrest and apoptosis. Furthermore, miR­873 was downregulated while Bcl-2 was upregulated in the tissues of twelve high-grade glioma patients compared to seven normal brain tissues, and the miR­873 level was negatively correlated with the Bcl-2 protein level. A luciferase reporter assay further confirmed that Bcl-2 was a direct target of miR­873, and miR­873 decreased the level of the Bcl-2 protein in cisplatin-resistant glioma cells. Notably, re-expression of Bcl-2 attenuated the function of miR­873 in cisplatin-resistant glioma cells and the sensitivity of the cells to cisplatin. Taken together, these data suggest that miR­873 might be a potential marker for cisplatin resistance and a promising sensitizer in cisplatin treatment.


Subject(s)
Brain Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Antineoplastic Agents/pharmacology , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cisplatin/pharmacology , Glioma/pathology , Humans , MicroRNAs , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
Oncol Rep ; 31(3): 1445-52, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24378652

ABSTRACT

microRNA (miRNA) dysregulation is associated with various types of human cancer by regulating cancer cell survival, proliferation and invasion. Aberrant expression of microRNA-503 (miR-503) has been reported in several cancer profiles. However, potential linkage of miR-503 levels and the underlying regulatory mechanisms in human glioblastoma multiforme (GBM) remain unclear. In the present study, we showed for the first time that the expression of miR-503 was significantly reduced in GBM tissues and cell lines (U251 and U87MG) relative to normal brain tissues. Furthermore, our results demonstrated that overexpression of miR-503 in GBM cell lines not only suppressed cell proliferation through inducing G0/G1 cell cycle arrest and apoptosis, but also inhibited cancer cell migration and tumor invasion. In addition, we identified insulin-like growth factor-1 (IGF­1R) receptor mRNA is a bona fide target of miR-503 by computational analysis followed by luciferase reporter assays. Of note, upregulation of miR-503 in GBM cells suppressed endogenous IGF-1R protein expression. Further mechanistic analysis revealed that forced expression of miR-503 inhibited AKT activation, suggesting the tumor suppressive effect of miR-503 in GBM cells is partially mediated by phosphatidylinositol 3-kinase/AKT signaling. Taken together, the results of the present study demonstrated that miR-503 is a tumor suppressor for GBM and a favorable factor against glioma progression through targeting IGF-1R, thus providing a new evidence-supported prognostic marker for GBM diagnosis.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Receptor, IGF Type 1/genetics , Apoptosis , Brain Neoplasms , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , G1 Phase Cell Cycle Checkpoints , Glioblastoma , Humans , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, IGF Type 1/metabolism
9.
Asian Pac J Cancer Prev ; 15(23): 10181-5, 2014.
Article in English | MEDLINE | ID: mdl-25556445

ABSTRACT

BACKGROUND: MicroRNAs, small noncoding RNA molecules, can regulate mammalian cell growth, apoptosis and differentiation by controlling the expression of target genes. The aim of this study was to investigate the function of miR-323-5p in the glioma cell line, U251. MATERIALS AND METHODS: After over-expression of miR-323- 5p using miR-323-5p mimics, cell growth, apoptosis and migration were tested by MTT, flow cytometry and cell wound healing assay, respectively. We also assessed the influence of miR-323-5p on the mRNA expression of IGF- 1R by quantitative real-time reverse transcriptase PCR (qRT-PCR), and on the protein levels by Western blot analysi. In addition, dual-luciferase reporter assays were performed to determine the target site of miR-323-5p to IGF-1R 3'UTR. RESULTS: Our findings showed that over-expression of miR-323-5p could promote apoptosis of U251 and inhibit the proliferation and migration of the glioma cells. CONCLUSIONS: This study demonstrated that increased expression of miR-323-5p might be related to glioma progression, which indicates a potential role of miR-323-5p for clinical therapy.


Subject(s)
Apoptosis/genetics , Glioma/genetics , MicroRNAs/genetics , RNA, Messenger/metabolism , Receptors, Somatomedin/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Glioma/metabolism , Humans , MicroRNAs/metabolism , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...