Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2695: 121-126, 2023.
Article in English | MEDLINE | ID: mdl-37450115

ABSTRACT

Glioma is the most common intracranial malignant tumor. Over the past several years, liquid biopsy in diagnosis and treatment of solid tumors have made many progressions, but there is still a gap from a large clinical application of liquid biopsy in glioma due to many limitations. However, in recent years, researchers have made many explorations into liquid biopsy in glioma. In the future, the liquid biopsy of glioma, especially cerebrospinal fluid, will have a broad prospect. In this review, we will discuss the current research progressions of CSF biopsy in glioma in recent years.


Subject(s)
Brain Neoplasms , Glioma , Humans , Biomarkers, Tumor , Glioma/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/cerebrospinal fluid , Biopsy/methods , Liquid Biopsy/methods
2.
Cancer Res ; 82(22): 4234-4246, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36112059

ABSTRACT

MutS protein homolog 2 (MSH2) is a key element involved in the DNA mismatch repair (MMR) system, which is responsible for recognizing and repairing mispaired bases. Simultaneously, MSH2 identifies DNA adducts induced by temozolomide (TMZ) and triggers apoptosis and autophagy in tumor cells. Previous work has revealed that reduced MSH2 expression is often observed in patients with glioblastoma (GBM) who relapse after chemotherapy. Elucidation of the mechanism behind TMZ-mediated reduction of MSH2 could help improve GBM treatment. Here, we report significant upregulation of Mex-3 RNA binding family member A (MEX3A) in GBM tissues and cell lines following TMZ treatment. MEX3A bound to the MEX3 recognition element (MRE) of MSH2 mRNA, which in turn recruited CCR4-NOT complexes to target MSH2 mRNA for deadenylation and degradation. In addition, ectopic expression of MEX3A significantly decreased cellular DNA MMR activities and reduced the chemosensitivity of GBM cells via downregulation of MSH2, while depletion of MEX3A sensitized GBM cells to TMZ. In MGMT-deficient patients with GBM, MEX3A expression correlated with MSH2 levels, and high MEX3A expression was associated with poor prognosis. Overall, these findings reveal a potential mechanism by which MSH2 expression is reduced in post-TMZ recurrent GBM. SIGNIFICANCE: A MEX3A/CCR4-NOT/MSH2 axis plays a crucial role in promoting temozolomide resistance, providing new insights into the function of MEX3A and suggesting MEX3A as a potential therapeutic target in therapy-resistant glioblastoma.


Subject(s)
Antineoplastic Agents, Alkylating , Brain Neoplasms , DNA Mismatch Repair , Drug Resistance, Neoplasm , Glioblastoma , MutS Homolog 2 Protein , Temozolomide , Humans , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Neoplasm Recurrence, Local/drug therapy , RNA, Messenger , Temozolomide/pharmacology , Temozolomide/therapeutic use , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
Technol Cancer Res Treat ; 20: 15330338211034270, 2021.
Article in English | MEDLINE | ID: mdl-34269120

ABSTRACT

Aberrant expression of lymphoid enhancer-binding factor-1 (LEF1) has been identified in various hematological malignancies including multiple myeloma (MM). However, the exact role of LEF1 in MM remains largely unknown. Here, we showed that knockdown of LEF1 could apparently impair the proliferation, induce apoptosis and promote the ROS production in MM cell lines, suggesting that LEF1 might be involved in maintaining MM cell growth and survival. Moreover, we observed that the mRNA level of the deubiquitinase cylindromatosis (CYLD), a well-recognized tumor suppressor in MM, was significantly increased following LEF1 depletion in myeloma cells. Further study showed that LEF1 could directly associate with the promoter of CYLD gene and thus repress its transcription in MM cells. Intriguingly, LEF1 depletion-mediated CYLD upregulation was sufficient to negatively modulate NF-κB signaling pathway in MM cells. Moreover, the decrease in NF-κB activity following LEF1 knockdown could be largely rescued when CYLD was silenced in MM cells. Taken together, our study provided the compelling evidence to show that LEF1 may augment the proliferation and survival of MM cells through direct repression of CYLD transcription and subsequent activation of NF-κB signaling pathway, corroborating that LEF1 may become a potential therapeutic target against MM.


Subject(s)
Deubiquitinating Enzyme CYLD/metabolism , Gene Expression Regulation, Neoplastic , Lymphoid Enhancer-Binding Factor 1/genetics , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Signal Transduction , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Humans , Immunophenotyping , Lymphoid Enhancer-Binding Factor 1/metabolism , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Prognosis
5.
Neuro Oncol ; 23(3): 435-446, 2021 03 25.
Article in English | MEDLINE | ID: mdl-32813021

ABSTRACT

BACKGROUND: Our previous studies have indicated that miR-198 reduces cellular methylguanine DNA methyltransferase (MGMT) levels to enhance temozolomide sensitivity. Transforming growth factor beta 1 (TGF-ß1) switches off miR-198 expression by repressing K-homology splicing regulatory protein (KSRP) expression in epidermal keratinocytes. However, the underlying role of TGF-ß1 in temozolomide resistance has remained unknown. METHODS: The distribution of KSRP was detected by western blotting and immunofluorescence. Microarray analysis was used to compare the levels of long noncoding RNAs (lncRNAs) between TGF-ß1-treated and untreated cells. RNA immunoprecipitation was performed to verify the relationship between RNAs and KSRP. Flow cytometry and orthotopic and subcutaneous xenograft tumor models were used to determine the function of TGF-ß1 in temozolomide resistance. RESULTS: Overexpression of TGF-ß1 contributed to temozolomide resistance in MGMT promoter hypomethylated glioblastoma cells in vitro and in vivo. TGF-ß1 treatment reduced cellular MGMT levels through suppressing the expression of miR-198. However, TGF-ß1 upregulation did not affect KSRP expression in glioma cells. We identified and characterized 2 lncRNAs (H19 and HOXD-AS2) that were upregulated by TGF-ß1 through Smad signaling. H19 and HOXD-AS2 exhibited competitive binding to KSRP and prevented KSRP from binding to primary miR-198, thus decreasing miR-198 expression. HOXD-AS2 or H19 upregulation strongly promoted temozolomide resistance and MGMT expression. Moreover, KSRP depletion abrogated the effects of TGF-ß1 and lncRNAs on miR-198 and MGMT. Finally, we found that patients with low levels of TGF-ß1 or lncRNA expression benefited from temozolomide therapy. CONCLUSIONS: Our results reveal an underlying mechanism by which TGF-ß1 confers temozolomide resistance. Furthermore, our findings suggest that a novel combination of temozolomide with a TGF-ß inhibitor may serve as an effective therapy for glioblastomas.


Subject(s)
Glioblastoma , MicroRNAs , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Tumor , DNA Modification Methylases/genetics , DNA Repair Enzymes , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , MicroRNAs/genetics , Temozolomide/pharmacology , Temozolomide/therapeutic use , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/therapeutic use , Tumor Suppressor Proteins/genetics
6.
Biosci Rep ; 40(10)2020 10 30.
Article in English | MEDLINE | ID: mdl-32969473

ABSTRACT

The present study was to determine the roles of Angiotensin (Ang) II in the growth of lymphoma in nude mice and the proliferation and viability of the human Natural Killer/T (NK/T)-cell lymphoma cell line SNK-6, and the activation of downstream signaling pathway. Lymphoma samples and corresponding normal tissues were obtained from lymphoma patients. Proliferation of SNK-6 cells was detected by CCK8 or MTT assay. The levels of Ang II and its receptor Ang II type 1 receptor (AT1R) were higher in lymphoma tissues than those in control tissues. Ang II increased the lymphoma volume and size in nude mice, the proliferation and viability and the proliferating cell nuclear antigen (PCNA) and Ki67 levels of SNK-6 cells. Losartan, an antagonist of AT1R, reduced lymphoma volume and size in nude mice, and the proliferation and viability and the PCNA and Ki67 levels of SNK-6 cells. The levels of phosphorylated phosphatidylinositol 3-kinase (p-PI3K) and phosphorylated protein kinase B (p-Akt) were increased by Ang II and then reduced by losartan in SNK-6 cells. The proliferation and viability of SNK-6 cells were increased by Ang II, but these increases were inhibited by PI3K inhibitor wortmannin and Akt inhibitor MK2206. The increases of PCNA and Ki67 induced by Ang II were inhibited by wortmannin or MK2206 in SNK-6 cells. These results indicate that Ang II/AT1R is activated in lymphoma, and Ang II promotes the progression of lymphoma in nude mice and the proliferation and viability of SNK-6 cells via activating PI3K/Akt signaling pathway.


Subject(s)
Angiotensin II/metabolism , Lymphoma, Extranodal NK-T-Cell/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Aged , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Losartan/pharmacology , Losartan/therapeutic use , Lymph Nodes/pathology , Lymphoma, Extranodal NK-T-Cell/drug therapy , Male , Middle Aged , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor, Angiotensin, Type 1/analysis , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Wortmannin/pharmacology , Wortmannin/therapeutic use , Xenograft Model Antitumor Assays
7.
Bosn J Basic Med Sci ; 20(2): 209-217, 2020 May 01.
Article in English | MEDLINE | ID: mdl-31538911

ABSTRACT

Multiple myeloma (MM) is a hematologic cancer arising from plasma cells. Mesenchymal stem cells (MSCs) are a heterogeneous cell population in the bone marrow microenvironment. In this study, we evaluated the regulatory effects of MSCs on the invasion and drug resistance of MM cells U266 and LP-1. Bone marrow samples from MM patients and healthy subjects were collected. MSCs were extracted from bone marrow and cultured, and their phenotypes were identified by flow cytometry. The level of CXCL13 in the supernatant of cultured MSCs was detected by ELISA. The protein expression of CXCR5 (a specific receptor of CXCL13) in U266 and LP-1 cells was detected by Western blot. The effects of MSCs on the invasion of U266 and LP-1 cells and the resistance to bortezomib were assessed by Transwell and CCK-8 assay, respectively. The mRNA and protein expressions of BTK, NF-κB, BCL-2, and MDR-1 were detected by RT-PCR and Western blot, respectively. CXCL13 was secreted by MSCs in the bone marrow microenvironment, and the level in MSCs from MM patients was significantly higher than that of healthy subjects. CXCR5 was expressed in both U266 and LP-1 cells. The resistance of MM cells to bortezomib was enhanced by MSCs through CXCL13 secretion. The invasion and proliferation of U266 and LP-1 cells were promoted, and the mRNA and protein expressions of BTK, NF-κB, BCL-2, and MDR-1 were upregulated by MSCs. The basic biological functions of MM cells U266 and LP-1 were affected by MSCs via the CXCL13-mediated signaling pathway. This study provides valuable experimental evidence for clinical MM therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Chemokine CXCL13/metabolism , Mesenchymal Stem Cells/physiology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Adult , Aged , Case-Control Studies , Cell Culture Techniques , Cell Line, Tumor , Coculture Techniques , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Neoplasm Invasiveness
8.
J Neurooncol ; 143(2): 221-229, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30997639

ABSTRACT

PURPOSE: Rap2B, a member of the GTP-binding proteins, is generally up-regulated in numerous types of tumors. Nevertheless, the influence and regulatory mechanisms of Rap2B in gliomas are still not corroborated. Therefore, we analyzed the expression of Rap2B in glioma tissues and cells, and researched its significance in adhesion, proliferation, migration and invasion of the glioma cell line. METHODS: We analyzed the expression of Rap2B in different pathologic grades of glioma tissues by tissue microarray and immunohistochemistry. We assessed the expression of Rap2B in glioma tissue and non-tumor tissue by Western blot. And the expression of Rap2b protein in glioma cells and normal human astrocytes (NHA) was detected by Western blot. In addition, we disclosed the effect of Rap2B knockdown on cell adhesion, proliferation, migration and invasion by using cell attachment assay, CCK-8 assay, cell migration assay and Wound Healing assay, cell invasion assay, respectively. Western blot was used to detect the changes of expression level of NF-kB, MMP-2 and MMP-9 protein when downregulated the expression of Rap2B. RESULTS: The tissue microarray immunohistochemical results of glioma showed that the expression of Rap2B had no significant correlations between Rap2B expression and the clinicopathologic variables, including patient age (P = 0.352), gender (P = 0.858), WHO Grade (P = 0.693) and histology type (P = 0.877). Western blot analysis showed that the glioma tissue had a dramatically increase of Rap2B expression compared with the non-tumor tissues (P < 0.01). And the expression of Rap2B was markedly up-regulated in all 5 glioma cell lines compared with that in normal human astrocytes (NHA) (P < 0.01). We found that the ability of adhesion, proliferation, migration and invasion of glioma cells were significantly decreased after downregulated Rap2B expression compared with the control group (P < 0.05). In addition, Western blot results showed that the expression levels of NF-kB, MMP-2 and MMP-9 in the interference group were significantly lower than those in the negative control group (P < 0.05). CONCLUSIONS: Rap2B expression is up-regulated in glioma tissues and glioma cell lines. Knockdown of Rap2B inhibits glioma cells' adhesion and proliferation in vitro. Knockdown of Rap2B inhibits glioma cells' migration in vitro. Knockdown of Rap2B inhibits glioma cells' invasion and MMPs activity through NF-kB pathway.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Glioma/pathology , rap GTP-Binding Proteins/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Humans , Male , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , Prognosis , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured , rap GTP-Binding Proteins/antagonists & inhibitors , rap GTP-Binding Proteins/genetics
9.
Invest New Drugs ; 37(6): 1177-1186, 2019 12.
Article in English | MEDLINE | ID: mdl-30847721

ABSTRACT

The dyskeratosis congenita 1 (DKC1) gene is located on the X chromosome at Xq28. Dyskerin encoded by the DKC1 gene is associated with the formation of certain small RNAs and the telomerase activity. Inherited mutations in DKC1 inactivate the dyskerin and causes dyskeratosis congenital, which is characterized by skin defects, hematopoiesis failure, and increased susceptibility to cancer. DKC1 reportedly up-regulates in several human cancers, including renal cell carcinoma and prostate cancer. Dyskerin is deregulated in B-chronic lymphocytic leukemia and breast carcinomas, but its expression and function in glioma have hardly been investigated. Hence, we were prompted to collect tissue samples and implement cell experiments. Our study reveals that DKC1 expression is significantly increased in the pathological tissues of glioma compared with that in normal tissues. The increased staining of DKC1 is related to the World Health Organization stages of tumors. DKC1 knockdown also significantly inhibits glioma cell growth by altering the expression of cell cycle-relative molecules to arrest at the G1 phase. In the transwell chamber, DKC1 knockdown glioma cells exhibit low motility. Consistent with classic oncogenic pathways, N-cadherin, HIF-1α, and MMP2 expression levels are lower compared with those of the control group. Therefore, DKC1 up-regulation in gliomas is common and necessary for extensive tumor growth. The phenotype of glioma cell lines after DKC1 down-regulation suggests its use as a valuable clinical treatment strategy.


Subject(s)
Brain Neoplasms , Cell Cycle Proteins , Glioma , Nuclear Proteins , Adult , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Glioma/genetics , Glioma/metabolism , Glioma/mortality , Glioma/pathology , Humans , Kaplan-Meier Estimate , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
10.
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
11.
Oncogene ; 38(15): 2706-2721, 2019 04.
Article in English | MEDLINE | ID: mdl-30542120

ABSTRACT

Temozolomide was recognized as the first-line therapy for glioblastoma to prolong the survival of patients noticeably, while recent clinical studies found that some patients were not sensitive to temozolomide treatment. The possible mechanisms seemed to be methylguanine-DNA-methyltransferase (MGMT), mismatch repair, PARP, etc. And the abnormal expression of MGMT might be the most direct factor. In this study, we provide evidence that Fstl1 plays a vital role in temozolomide resistance by sequentially regulating DIP2A protein distribution, H3K9 acetylation (H3K9Ac), and MGMT transcription. As a multifunctional protein widely distributed in cells, DIP2A cooperates with the HDAC2-DMAP1 complex to enhance H3K9Ac deacetylation, prevent MGMT transcription, and increase temozolomide sensitivity. Fstl1, a glycoprotein highly expressed in glioblastoma, competitively binds DIP2A to block DIP2A nuclear translocation, so as to hinder DIP2A from binding the HDAC2-DMAP1 complex. The overexpression of Fstl1 promoted the expression of MGMT in association with increased promoter H3K9Ac. Upregulation of Fstl1 enhanced temozolomide resistance, whereas Fstl1 silencing obviously sensitized GBM cells to temozolomide both in vivo and in vitro. Moreover, DIP2A depletion abolished the effects of Fstl1 on MGMT expression and temozolomide resistance. These findings highlight an important role of Fstl1 in the regulation of temozolomide resistance by modulation of DIP2A/MGMT signaling.


Subject(s)
Carrier Proteins/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Follistatin-Related Proteins/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Nuclear Proteins/genetics , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Acetylation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Histone Deacetylase 2 , Humans , Promoter Regions, Genetic/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Transcription, Genetic/genetics , Up-Regulation/genetics
12.
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.

13.
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
14.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...