Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 13: 1214413, 2023.
Article in English | MEDLINE | ID: mdl-37771444

ABSTRACT

Background: Studies have shown that the immune infiltration of tumor microenvironment is related to the prognosis of glioblastoma, which is characterized by high heterogeneity, high recurrence rate and low survival rate. To unravel the role of ß1,3-N-acetylglucosaminyltransferase-9 (ß3GNT9) in the progression of glioblastoma, this study identifies the value of ß3GNT9 as a prognostic biomarker in glioblastoma, and investigates the relationship between ß3GNT9 expression and glioblastoma immune infiltration, migration and invasion. Methods: ß3GNT9 expression in glioblastoma was analyzed using the GEPIA database. The clinical features of glioblastoma were screened out from the TCGA database. The relationship between ß3GNT9 expression and clinical features was analyzed. The relationship between ß3GNT9 and the prognosis of glioblastoma was evaluated through univariate and multivariate COX regression analyses, and the survival analysis was conducted using the Kaplan-Meier method. GSEA was employed to predict the signaling pathway of ß3GNT9 in glioblastoma. The correlation between ß3GNT9 and tumor immune infiltration was analyzed using the related modules of CIBERSORT and TIMER. A172, U87MG and U251 cell lines were selected to verify ß3GNT9 expression in vitro. The effects of ß3GNT9 on the migration and invasion of glioblastoma were investigated through cell scratch and invasion assays. Results: ß3GNT9 expression in glioblastoma group was significantly higher than that in normal brain tissue group (P<0.05). The overall survival rate in high ß3GNT9 expression group was significantly lower than that in low ß3GNT9 expression group (P<0.05). Regression analyses suggested that ß3GNT9, involved primarily in glucosamine degradation and extracellular matrix receptor interaction, could be an independent prognostic factor for glioblastoma. CIBERSORT and GEPIA database analyses showed that ß3GNT9 was correlated with tumor infiltrating immune cells such as T follicular helper cells, activating natural killer cells, monocytes, macrophages, and eosinophils, thus affecting the immune microenvironment of glioblastoma. Cell experiments confirmed that ß3GNT9 was highly expressed in A172, U87MG and U251 cell lines (P<0.05), and promoted the migration and invasion of glioblastoma (P<0.05). Conclusion: The increased expression of ß3GNT9 in glioblastoma can affect the immune microenvironment of glioblastoma and promote its migration and invasion. ß3GNT9 can be used as a potential independent prognostic biomarker for patients with glioblastoma.

2.
Med Mol Morphol ; 52(2): 114-122, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30564876

ABSTRACT

This study aimed to investigate the function of glioma stem cells (GSCs) and the role of PCAT1. This study dissociated the differences between GSCs and glioma cells in terms of apoptosis rate and γH2AX positive cells levels after radiation. Microarray was carried out to detect that expressed PCAT1, and it was testified by RT-qPCR. After transfection, GSCs were used to investigate the influence of PCAT1 on radiation sensitivity. Sphere-formation capability was first examined. Cell apoptosis rate after radiation of 0 Gy or 6 Gy was analyzed by flow cytometry, and the level of γH2AX positive cells after 6 Gy radiation were compared. CCK8 assay was used to investigate the cell proliferation and RT-qPCR was used to examine miR-129-5p and HMGB1 expression. GSCs exhibited great capability in sphere formation and lower expression in apoptosis and γH2AX positive cells rates after 6 Gy radiation. PCAT1 had higher expression in GSCs. PCAT1 knockdown restrained the sphere-formation ability, increased the apoptosis rate and DNA damage under the treatment of radiation. Moreover, knockdown of PCAT1 inhibited the cell proliferation. In addition, silencing PCAT1 could increase the expression of miR-129-5p and decrease the expression of HMGB1. PCAT1 was overexpressed in GSCs and played a facilitating role in radiation resistance.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Neoplastic Stem Cells/radiation effects , RNA, Long Noncoding/genetics , Radiation Tolerance/genetics , Apoptosis/genetics , Apoptosis/radiation effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/radiation effects , Glioma/pathology , Humans , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/radiation effects
3.
DNA Cell Biol ; 37(11): 917-924, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30388035

ABSTRACT

As a novel type of noncoding RNAs, circular RNAs (circRNAs) have been acknowledged as pivot effectors in multiple physiological and pathological processes, including cancer. Although an increasing number of circRNAs are gradually discovered, their function and mechanism in tumorigenesis remain largely unknown. In this study, we identified hsa_circ_0074362 as an oncogene in glioma. We examined hsa_circ_0074362 expression in glioma tissues and cell lines by qRT-PCR and investigated its functions by using CCK8 and transwell assays. Luciferase reporter assays were conducted to confirm the interaction between miR-1236-3p and hsa_circ_0074362 or HOXB7. Results demonstrated that hsa_circ_0074362 was significantly upregulated in glioma tissues compared with normal tissues, and hsa_circ_0074362 overexpression was correlated with clinical severity and poor prognosis in patients with glioma. Moreover, hsa_circ_0074362 knockdown remarkably suppressed glioma cell proliferation, migration, and invasion. Luciferase reporter assay indicated that hsa_circ_0074362 acted as a sponge of miR-1236-3p to promote HOXB7 expression. Thus, hsa_circ_0074362 played a crucial role in glioma progression by regulating the miR-1236-3p/HOXB7 pathway.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioma/genetics , Homeodomain Proteins/genetics , MicroRNAs/genetics , RNA/genetics , Adult , Animals , Base Sequence , Binding Sites , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Glioma/metabolism , Glioma/pathology , Glioma/therapy , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Nude , MicroRNAs/metabolism , Middle Aged , Neoplasm Transplantation , Neuroglia/metabolism , Neuroglia/pathology , RNA/metabolism , RNA, Circular , Signal Transduction
4.
DNA Cell Biol ; 37(9): 791-797, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30010402

ABSTRACT

As a new member of the noncoding RNA family, circular RNAs (circRNAs) have been demonstrated as critical regulators in various physiological and pathological processes, such as tumorigenesis. However, the role of circRNAs has not been well understood until now. In our study, we found that circRNA hsa_circ_0000177 was upregulated in glioma tissues and cell lines. Also, hsa_circ_0000177 overexpression was associated with poor prognosis in glioma patients. Through functional experiments, we found that hsa_circ_0000177 knockdown dramatically inhibited glioma cell proliferation and invasion in vitro. Consistently, hsa_circ_0000177 knockdown significantly repressed glioma growth in vivo. In terms of mechanism, we used bioinformatics analysis and identified hsa_circ_0000177 as a miR-638 sponge. We showed that miR-638 inhibition could restore the proliferation and invasion of glioma cells transfected with hsa_circ_0000177 small interfering RNA. Furthermore, we demonstrated that frizzled class receptor 7 (FZD7) was targeted by miR-638 and upregulated by hsa_circ_0000177. Through upregulating FZD7 expression, hsa_circ_0000177 activated Wnt signaling and facilitated glioma growth. Taken together, our study revealed a novel signaling pathway involved in glioma progression.


Subject(s)
Frizzled Receptors/metabolism , Gene Expression Regulation, Neoplastic , Glioma/pathology , MicroRNAs/genetics , RNA/genetics , Wnt Signaling Pathway , Animals , Cell Line, Tumor , Cell Proliferation , Computational Biology , Disease Progression , Female , Frizzled Receptors/antagonists & inhibitors , Frizzled Receptors/genetics , Glioma/genetics , Glioma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , RNA, Circular , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...