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
1.
Invest New Drugs ; 37(4): 646-657, 2019 08.
Article in English | MEDLINE | ID: mdl-30328556

ABSTRACT

Recently, XRCC1 polymorphisms were reported to be associated with glioma in Chinese population. However, only a few studies reported on the XRCC1 expression, and cancer progression. In this study, we investigated whether XRCC1 plays a role in glioma pathogenesis. Using the tissue microarray technology, we found that XRCC1 expression is significantly decreased in glioma compared with tumor adjacent normal brain tissue (P < 0.01, χ2 test) and reduced XRCC1 staining was associated with WHO stages (P < 0.05, χ2 test). The mRNA and protein levels of XRCC1 were significantly downregulated in human primary glioma tissues (P < 0.001, χ2 test). We also found that XRCC1 was significantly decreased in glioma cell lines compared to normal human astrocytes (P < 0.01, χ2 test). Overexpression of XRCC1 dramatically reduced the proliferation and caused cessation of cell cycle. The reduced cell proliferation is due to G1 phase arrest as cyclin D1 is diminished whereas p16 is upregulated. We further demonstrated that XRCC1 overexpression suppressed the glioma cell migration and invasion abilities by targeting MMP-2. In addition, we also found that overexpression of XRCC1 sharply inhibited angiogenesis, which correlated with down-regulation of VEGF. The data indicate that XRCC1 may be a tumor suppressor involved in the progression of glioma.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , X-ray Repair Cross Complementing Protein 1/genetics , Astrocytes/metabolism , Brain/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Glioma/metabolism , Glioma/pathology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Matrix Metalloproteinase 2/metabolism , Middle Aged , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/metabolism , Wound Healing , X-ray Repair Cross Complementing Protein 1/metabolism
2.
Br J Cancer ; 118(4): 522-533, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29348487

ABSTRACT

BACKGROUND: The therapeutic resistance to ionising radiation (IR) and anti-angiogenesis mainly impair the prognosis of patients with glioblastoma. The primary and secondary MET aberrant activation is one crucial factor for these resistances. The kringle 1 domain of hepatocyte growth factor (HGFK1), an angiogenic inhibitor, contains a high-affinity binding domain of MET; however, its effects on glioblastoma remain elusive. METHODS: We formed the nanoparticles consisting of a folate receptor-targeted nanoparticle-mediated HGFK1 gene (H1/pHGFK1) and studied its anti-tumoural and radiosensitive activities in both subcutaneous and orthotopic human glioma cell-xenografted mouse models. We then elucidated its molecular mechanisms in human glioblastoma cell lines in vitro. RESULTS: We demonstrated for the first time that peritumoural injection of H1/pHGFK1 nanoparticles significantly inhibited tumour growth and prolonged survival in tumour-bearing mice, as well as enhanced the anti-tumoural efficacies of IR in vivo by reducing Ki-67 expression, enhancing TUNEL staining-indicated apoptotic indexes, reducing microvascular intensity and reversing IR-induced MET overexpression in tumour tissues. Furthermore, we showed that HGFK1 suppressed the proliferation and induced cell apoptosis and enhanced sensitivity to IR in glioblastoma cell lines, mainly by suppressing the activities of MET receptor, down-regulating ATM-Chk2 axis but up-regulating Chk1. CONCLUSIONS: H1/pHGFK1 exerts anti-tumoural and radiosensitive activities mainly through the inhibition and reversal of IR-induced MET and ATM-Chk2 axis activities in glioblastoma. H1/pHGFK1 nanoparticles are a potential radiosensitiser and angiogenic inhibitor for glioblastoma treatment.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Hepatocyte Growth Factor/genetics , Plasmids/administration & dosage , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Radiation-Sensitizing Agents/administration & dosage , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/genetics , Hepatocyte Growth Factor/chemistry , Humans , Kringles , Mice , Nanoparticles/administration & dosage , Plasmids/genetics , Radiation-Sensitizing Agents/pharmacology , Xenograft Model Antitumor Assays
3.
Int J Clin Exp Pathol ; 10(9): 9318-9329, 2017.
Article in English | MEDLINE | ID: mdl-31966804

ABSTRACT

Recently, collagen triple helix repeat containing-1 (CTHRC1) has been reported to be increased in several types of human solid cancers and to be associated with tumor invasion and metastasis. However, the expression and function of CTHRC1 in glioma have not yet been reported. In the present study, we investigated whether CTHRC1 plays a role in glioma pathogenesis. Using the tissue microarray technology, we found that CTHRC1 expression is significantly increased in glioma compared with tumor adjacent normal brain tissue (P<0.01, χ2 test) and increased CTHRC1 staining was associated with WHO stages (P<0.05, χ2 test). The mRNA and protein levels of CTHRC1 were significantly upregulated in human primary glioma tissues (P<0.001, χ2 test). We also found that CTHRC1 was significantly increased in glioma cell lines compared to normal human astrocytes (P<0.01, χ2 test). Furthermore, Knockdown of CTHRC1 suppressed glioma cell invasion and inhibited enzyme activity of MMP-2. Moreover, our data showed that knockdown of CTHRC1 inhibited glioma cell migration and adhesion capacity when compared with the control cells, and CTHRC1-siRNA reduced the levels of phosphorylated Src and FAK protein expression. Taken together, this study suggests that CTHRC1 plays a role in glioma development and progression by regulating invasion, migration and adhesion capabilities of cancer cells.

4.
Int J Clin Exp Pathol ; 8(6): 6952-9, 2015.
Article in English | MEDLINE | ID: mdl-26261583

ABSTRACT

PinX1, a conserved nuclear protein, could maintain telomere integrity and plays an important role in regulating telomerase activity. It has been reported that the expression of PinX1 is down-regulated in some cancer and associated with cancer prognosis. However, the value of PinX1 in gliomas has not been studied. In this study, two independent retrospective gliomas cohorts with the corresponding gliomas tissue microarrays (TMAs) were established to detect the expression level of PinX1 and the correlation of PinX1 expression with the clinicopathological features and the patients' survival. Compared with non-cancerous brain tissues, PinX1 protein levels were remarkably up-regulated in gliomas (P = 0.001), and further increased from benign gliomas tissues to malignant gliomas tissues (P = 0.090). Moreover, high PinX1 expression was significantly positively associated with gliomas WHO grade in the training set (P = 0.019) and the validation set (P = 0.037). High PinX1 expression significantly correlated with a worse 5-year overall (P = 0.016) and disease-specific survival (P = 0.026). Simultaneously, the multivariate COX regression analysis showed that PinX1 was an independent unfavorable prognostic factor for 5-year overall survival (hazard ratio (HR) = 2.078, P = 0.015) and disease-specific survival (HR = 2.429, P = 0.012) after adjusting with age, sex and WHO grade in gliomas. In conclusion, PinX1 expression may serve as a prognostic and predictive biomarker for gliomas.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/chemistry , Glioma/chemistry , Tumor Suppressor Proteins/analysis , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Cycle Proteins , Chi-Square Distribution , Disease-Free Survival , Female , Glioma/mortality , Glioma/pathology , Glioma/therapy , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Predictive Value of Tests , Proportional Hazards Models , Reproducibility of Results , Retrospective Studies , Risk Factors , Time Factors , Tissue Array Analysis , Treatment Outcome , Up-Regulation
5.
Med Oncol ; 32(3): 73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25698538

ABSTRACT

PinX1 induces apoptosis and suppresses cell proliferation in some cancer cells, and the expression of PinX1 is frequently decreased in some cancer and negatively associated with metastasis and prognosis. However, the precise roles of PinX1 in gliomas have not been studied. In this study, we found that PinX1 obviously reduced the gliomas cell proliferation through regulating the expressions of cell cycle-relative molecules to arrest cell at G1 phase and down-regulating the expression of component telomerase reverse transcriptase (hTERT in human), which is the hardcore of telomerase. Moreover, PinX1 could suppress the abilities of gliomas cell wound healing, migration and invasion via suppressing MMP-2 expression and increasing TIMP-2 expression. In conclusion, our results suggested that PinX1 may be a potential suppressive gene in the progression of gliomas.


Subject(s)
Glioma/genetics , Glioma/pathology , Tumor Suppressor Proteins/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 2/metabolism , Telomerase/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tumor Suppressor Proteins/metabolism
6.
Med Oncol ; 31(10): 227, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25201578

ABSTRACT

This study was designed to explore the role of Cullin1 (Cul1) in the pathogenesis of human glioma and to investigate the role of Cul1 in the growth, migration and invasion of glioma cells. Expression of Cul1 in 191 glioma tissues, 8 normal brain tissues and 8 tumor adjacent normal brain tissues was analyzed by tissue microarray and immunohistochemistry. Cul1 expression in human glioblastoma cells was knocked down by specific siRNA to study the effect of down-regulation of Cul1 on proliferation, invasion and migration of glioma cells. Our results showed that Cul1 expression increased significantly in tissues from the benign tumor and malignant tumor in comparison with those from the tumor-adjacent normal brain (P<0.05 for both). We did not find any correlation between Cul1 expression and clinicopathological parameters. In addition, we found that knockdown of Cul1 by RNA interference markedly inhibited cell proliferation and caused cessation of cell cycle. This reduced cell proliferation was due to G1 phase arrest as cyclinA, cyclinD1 and cyclinE were diminished, whereas p21 and p27 were up-regulated. We further demonstrated that silencing of Cul1 in glioma cells inhibited the cell migration and invasion abilities, and down-regulation of MMP-2 and MMP-9 expression greatly contributed to the reduced cell invasion and migration abilities. Our data indicated that Cul1 expression significantly increased in human glioma, and it may be involved in proliferation, migration and invasion of glioma cells.


Subject(s)
Brain Neoplasms/pathology , Cell Movement/physiology , Cell Proliferation/physiology , Cullin Proteins/physiology , Glioma/pathology , Blotting, Western , Brain Neoplasms/etiology , Cell Cycle Proteins/metabolism , Female , Gene Knockdown Techniques , Glioma/etiology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Invasiveness , RNA, Small Interfering/genetics , Tissue Array Analysis , Tumor Cells, Cultured
7.
J Neurooncol ; 115(2): 179-88, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23943502

ABSTRACT

Previous studies reported that miR-29c is significantly downregulated in several tumors. However, little is known about the effect and molecular mechanisms of action of miR-29c in human glioma. Using quantitative RT-PCR, we demonstrated that miR-29c was significantly downregulated in glioma cell lines and human primary glioma tissues, compared to normal human astrocytes and matched non-tumor associated tissues (P < 0.05, χ(2) test). Overexpression of miR-29c dramatically reduced the proliferation and caused cessation of cell cycle. The reduced cell proliferation is due to G1 phase arrest as cyclin D1 and cyclin E are diminished whereas p27 and p21 are upregulated. We further demonstrated that miR-29c overexpression suppressed the glioma cell migration and invasion abilities by targeting MMP-2. In addition, we also found that overexpression of miR-29c sharply inhibited angiogenesis, which correlated with down-regulation of VEGF. The data indicate that miR-29c may be a tumor suppressor involved in the progression of glioma.


Subject(s)
Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Glioma/pathology , MicroRNAs/genetics , Neovascularization, Pathologic , Apoptosis , Blotting, Western , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Glioma/blood supply , Glioma/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism
8.
J Cancer Res Clin Oncol ; 138(6): 991-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362300

ABSTRACT

PURPOSE: The purposes of our study were to elucidate the role of BRG1 in the development of human glioma and to determine the effect of BRG1 on glioma cell growth, migration and invasion. METHODS: Using tissue microarray and immunohistochemistry, we evaluated BRG1 staining in 190 glioma tissues, 8 normal brain tissues and 8 tumor adjacent normal brain tissues. We studied glioma cell proliferative ability with reduced BRG1 expression by siRNA using CCK-8 cell proliferation assay and cell cycle analysis. We studied the role of BRG1 in glioma cell migration and invasion by cell migration assay and matrigel invasion assay. We performed western blot to detect cyclin D1, cyclin B1 and MMP-2 protein expression. We also detected MMP-2 enzyme activity by gelatin zymography. RESULTS: Our results showed that BRG1 expression was increased in benign tumor and malignant tumor compared with tumor adjacent normal brain tissue (P < 0.01 for both). We did not find any correlation between BRG1 expression and clinicopathological parameters. In addition, we found that knockdown of BRG1 in glioma cell lines inhibits cell growth due to the G1 phase arrest by downregulating cyclin D1. We further demonstrated that silencing of BRG1 in glioma cells inhibited the cell migration and invasion abilities, and downregulation of MMP-2 expression greatly contributed to the reduced cell invasion and migration abilities. CONCLUSIONS: Our data indicated that BRG1 expression is significantly increased in human glioma and it may be involved in the process of glioma cell proliferation, migration and invasion.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Movement/physiology , DNA Helicases/biosynthesis , Glioma/metabolism , Glioma/pathology , Nuclear Proteins/biosynthesis , Transcription Factors/biosynthesis , Brain Neoplasms/genetics , Cell Cycle/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gene Knockdown Techniques/methods , Gene Silencing , Glioma/genetics , Humans , Immunohistochemistry/methods , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Middle Aged , Neoplasm Invasiveness , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , Sincalide/genetics , Sincalide/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
J Cancer Res Clin Oncol ; 137(12): 1823-30, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21922326

ABSTRACT

PURPOSE: The aim of this study is to investigate whether the expression of RUNX3 is related to the development of glioma, and the role of RUNX3 in glioma cells growth, invasion and migration. METHODS: We analyzed the protein expression of RUNX3 by immunohistochemistry in 188 glioma tissues, 8 normal brain tissues and 8 tumor adjacent normal brain tissues using tissue microarray technique. We studied whether RUNX3 restoration can suppress glioma cells growth, invasion and migration by performing MTT cell proliferation assay, matrigel cell invasion assay, wound-healing assay and migration assay. We also detected MMP-2 protein expression and enzyme activity by western blot analysis and gelatin zymography. RESULTS: We found that RUNX3 expression was decreased in benign tumor and malignant tumor compared with tumor adjacent normal brain tissue (P < 0.01 and P < 0.05, respectively). We did not find any correlation between RUNX3 expression and clinicopathological parameters. In addition, we demonstrated that re-expression of RUNX3 in glioma cells resulted in significantly inhibited cell invasion and migration abilities. This reduced cell invasion and migration abilities were due to MMP-2 protein expression and enzyme activity suppression after RUNX3 restoration. CONCLUSIONS: Our data indicated that RUNX3 expression is significantly decreased in human glioma, and targeting of the RUNX3 pathway may constitute a potential treatment modality for glioma.


Subject(s)
Brain Neoplasms/pathology , Core Binding Factor Alpha 3 Subunit/physiology , Glioma/pathology , Brain Neoplasms/chemistry , Cell Line, Tumor , Cell Movement , Core Binding Factor Alpha 3 Subunit/analysis , Glioma/chemistry , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors , Neoplasm Invasiveness
SELECTION OF CITATIONS
SEARCH DETAIL
...