1.Research progress on platelets in glioma.
Mingrong ZUO ; Tengfei LI ; Zhihao WANG ; Yufan XIANG ; Siliang CHEN ; Yanhui LIU
Chinese Medical Journal 2025;138(1):28-37
Gliomas are the most common primary neuroepithelial tumors of the central nervous system in adults, of which glioblastoma is the deadliest subtype. Apart from the intrinsically indestructible characteristics of glioma (stem) cells, accumulating evidence suggests that the tumor microenvironment also plays a vital role in the refractoriness of glioblastoma. The primary functions of platelets are to stop bleeding and regulate thrombosis under physiological conditions. Furthermore, platelets are also active elements that participate in a variety of processes of tumor development, including tumor growth, invasion, and chemoresistance. Glioma cells recruit and activate resting platelets to become tumor-educated platelets (TEPs), which in turn can promote the proliferation, invasion, stemness, and chemoresistance of glioma cells. TEPs can be used to obtain genetic information about gliomas, which is helpful for early diagnosis and monitoring of therapeutic effects. Platelet membranes are intriguing biomimetic materials for developing efficacious drug carriers to enhance antiglioma activity. Herein, we review the recent research referring to the contribution of platelets to the malignant characteristics of gliomas and focusing on the molecular mechanisms mediating the interaction between TEPs and glioma (stem) cells, as well as present the challenges and opportunities in targeting platelets for glioma therapy.
Humans
;
Glioma/metabolism*
;
Blood Platelets/physiology*
;
Brain Neoplasms/pathology*
;
Tumor Microenvironment
3.Sialyltransferase ST3GAL1 promotes malignant progression in glioma.
Zihao ZHAO ; Wenjing ZHENG ; Lingling ZHANG ; Wenjie SONG ; Tao WANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(4):308-317
Objective To investigate the clinical relevance and diagnostic or prognostic value of ST3β-galactoside α-2, 3-sialyltransferase 1 (ST3GAL1) in glioma and to confirm its role in promoting malignant phenotypes. Methods Using data from The Cancer Genome Atlas (TCGA) database, we analyzed the correlation between ST3GAL1 expression levels in glioma and clinical parameters to evaluate its diagnostic and prognostic value. The impact of ST3GAL1 on malignant phenotypes of glioma cells-including proliferation, cell cycle progression, apoptosis, and invasion was further validated through ST3GAL1 knockdown experiments. Results The expression level of ST3GAL1 was significantly higher in glioma tissues compared to healthy brain tissues and showed a strong correlation with clinical characteristics of glioma patients. Survival analysis and receiver operating characteristic (ROC) curve demonstrated that ST3GAL1 could serve as a potential diagnostic and prognostic biomarker for glioma. Knockdown of ST3GAL1 suppressed proliferation, invasion, and migration capabilities of glioma cell lines, and induced G1-phase cell cycle arrest. Conclusion ST3GAL1 promotes malignant phenotypes in glioma and plays a critical role in its malignant progression, suggesting its potential as a biomarker for glioma diagnosis and prognosis.
Humans
;
Sialyltransferases/metabolism*
;
Glioma/diagnosis*
;
Cell Proliferation/genetics*
;
Cell Line, Tumor
;
Brain Neoplasms/enzymology*
;
beta-Galactoside alpha-2,3-Sialyltransferase
;
Disease Progression
;
Prognosis
;
Cell Movement/genetics*
;
Apoptosis/genetics*
;
Male
;
Female
;
Gene Expression Regulation, Neoplastic
;
Biomarkers, Tumor/metabolism*
;
Middle Aged
4.Circ_EPHB4 regulates temozolomide sensitivity in glioma cells through the miR-424-5p/Wnt3 axis.
Yuxiang LIAO ; Jingping LIU ; Bo LIU ; Xiyun FEI ; Chen JIN
Journal of Southern Medical University 2025;45(5):942-953
OBJECTIVES:
To investigate the mechanism by which circ_EPHB4 regulates temozolomide (TMZ) sensitivity of glioma cells through the miR-424-5p/Wnt3 signal axis.
METHODS:
We detected the expression levels of circ_EPHB4, miR-424-5p and Wnt3 mRNA in glioma specimens from 25 patients with primary glioma and 25 patients experiencing relapse following temozolomide-based chemotherapy and in TMZ-sensitive and -resistant glioma A172 and SHG44 cells with circ_EPHB4 knockdown using qRT-PCR, and Wnt3 protein expression level was detected with Western blotting. Cell viability, colony-forming ability, and apoptosis of the cells with circ_EPHB4 knockdown were assessed, and the targeted regulation relationship between circ_EPHB4, miR-424-5p, and Wnt3 was verified by dual luciferase reporter assay and RNA immunoprecipitation (RIP) experiments. The effect of circ_EPHB4 knockdown on tumorigenesis of glioma cells was evaluated in subcutaneous tumor-bearing nude mouse models.
RESULTS:
The expression of circ_EPHB4 was significantly increased in glioma tissues and cells as compared with normal neural tissues and astrocytes (P=0.014). In TMZ-resistant glioma cells, circ_EPHB4 knockdown resulted in an obvious reduction of IC50 value of TMZ, inhibited cell colony formation, and promoted cell apoptosis, and these effects were reversed by miR-424-5p knockdown. The expressions of miR-424-5p and circ_EPHB4 were negatively correlated in glioma tissues (P=0.011). MiR-424-5p knockdown also attenuated the effect of circ_EPHB4 knockdown on expressions of PCNA, P-gp, MRP1 and bax.
CONCLUSIONS
Circ_EPHB4 regulates Wnt3 expression through "sponge adsorption" of miR-424-5p, thereby modulating TMZ-resistant glioblastoma cell clonogenesis, apoptosis, and TMZ sensitivity, suggesting the potential of circ_EPHB4 as a therapeutic target for reversing drug resistance of gliomas.
MicroRNAs/genetics*
;
Humans
;
Temozolomide
;
Glioma/genetics*
;
Animals
;
Mice, Nude
;
Cell Line, Tumor
;
Wnt3 Protein/metabolism*
;
Mice
;
Apoptosis
;
RNA, Circular
;
Drug Resistance, Neoplasm
;
Brain Neoplasms/pathology*
;
Signal Transduction
5.circ_EPHB4 synergizes with YTHDF3 to promote glioma progression via m6A-dependent stabilization of Wnt3.
Chen JIN ; Jingping LIU ; Bo LIU ; Xiyun FEI ; Yuxiang LIAO
Journal of Southern Medical University 2025;45(11):2320-2329
OBJECTIVES:
To investigate the oncogenic role of circular RNA circ_EPHB4 in glioma and its molecular mechanism.
METHODS:
Microarray analysis was performed to identify the differentially expressed circRNAs in glioma tissues. The effects of circ_EPHB4 on glioma cell migration, invasion and epithelial-mesenchymal transition (EMT) in vitro and tumorigenicity in vivo were assessed using scratch wound healing assay, Transwell invasion assay and nude mouse models bearing subcutaneous tumors. RNA immunoprecipitation (RIP), RNA stability assays, and gene overexpression and silencing techniques were employed to validate the synergistic regulatory effect of circ_EPHB4 and the N6-methyladenosine (m6A) reader protein YTHDF3 on Wnt3 expression.
RESULTS:
Circ_EPHB4 was significantly overexpressed by 2.3 folds (|log2FC|=1.2, P<0.01) in glioma tissues compared to the adjacent tissues, and by 2.5 folds in glioma cell line U373 compared to normal cells (P<0.001). Overexpression of circ_EPHB4 significantly enhanced migration and invasion of glioma cells, and promoted the expressions of EMT markers N-cadherin and vimentin. In the tumor-bearing mouse models, the tumor volume in circ_EPHB4 overexpression group was significantly greater than that in the control group, and the lung metastatic foci increased by 4.2 folds. Overexpression of circ_EPHB4 promoted oncogenesis by upregulating Wnt3 expression, while YTHDF3 extended the half-life of Wnt3 mRNA in an m6A-dependent manner. Simultaneous knockdown of circ_EPHB4 and YTHDF3 resulted in an obvious reduction of Wnt3 mRNA expression by up to 47% compared to its level following knocking down either circ_EPHB4 or YTHDF3 alone.
CONCLUSIONS
Circ_EPHB4 and YTHDF3 promote glioma progression by jointly targeting the Wnt3 signaling pathway, which may provide a new therapeutic strategy for gliomas.
Glioma/genetics*
;
Humans
;
Animals
;
Cell Line, Tumor
;
RNA-Binding Proteins/genetics*
;
RNA, Circular
;
Epithelial-Mesenchymal Transition
;
Mice, Nude
;
Cell Movement
;
Wnt3 Protein/genetics*
;
Mice
;
Disease Progression
;
Adenosine/metabolism*
;
Brain Neoplasms/metabolism*
;
Gene Expression Regulation, Neoplastic
6.Progress on the functions and mechanisms of natural products in anti-glioma therapy.
Yanting LI ; Shuhui QU ; Jiayi ZUO ; Haoping LONG ; Feng CAO ; Feng JIANG
Chinese Journal of Natural Medicines (English Ed.) 2025;23(5):541-559
Glioma, the most prevalent primary tumor of the central nervous system (CNS), is also the most lethal primary malignant tumor. Currently, there are limited chemotherapeutics available for glioma treatment, necessitating further research to identify and develop new chemotherapeutic agents. A significant approach to discovering anti-glioma drugs involves isolating antitumor active ingredients from natural products (NPs) and optimizing their structures. Additionally, targeted drug delivery systems (TDDSs) are employed to enhance drug solubility and stability and overcome the blood-brain barrier (BBB). TDDSs can penetrate deep into the brain, increase drug concentration and retention time in the CNS, and improve the targeting efficiency of NPs, thereby reducing adverse effects and enhancing anti-glioma efficacy. This paper reviews the research progress of anti-glioma activities of NPs, including alkaloids, polyphenols, flavonoids, terpenoids, saponins, quinones, and their synthetic derivatives over the past decade. The review also summarizes anti-glioma mechanisms, such as suppression of related protein expression, regulation of reactive oxygen species (ROS) levels, control of apoptosis signaling pathways, reduction of matrix metalloproteinases (MMPs) expression, blocking of vascular endothelial growth factor (VEGF), and reversal of immunosuppression. Furthermore, the functions and advantages of NP-based TDDSs in anti-glioma therapy are examined. The key information presented in this review will be valuable for the research and development of NP-based anti-glioma drugs and related TDDSs.
Humans
;
Glioma/metabolism*
;
Biological Products/therapeutic use*
;
Animals
;
Brain Neoplasms/genetics*
;
Drug Delivery Systems
;
Antineoplastic Agents/therapeutic use*
;
Blood-Brain Barrier/metabolism*
;
Apoptosis/drug effects*
7.Research progress in the small G-protein Rac1.
Yiheng YANG ; Shuling ZHAO ; Changyong LIANG
Chinese Journal of Biotechnology 2024;40(11):3902-3911
The small G-protein Rac1 is the main regulatory factor of the actin cytoskeleton. Rac1 cycles between the inactive GDP-bound form and the active GTP-bound form. Rac1 not only promotes viral replication and infection, but also regulates the actin cytoskeleton rearrangement, adhesion, and invasion of glioma cells. In addition, Rac1 is implicated in human diseases such as tumors and epilepsy. This article reviews the latest research on the small G-protein Rac1 in virology, cell biology, and human pathology. It is found that the existence of Rac1 is closely related to the replication and infection of viruses, that is, inhibiting the existence of Rac1 can effectively reduce the replication and transportation of viruses, providing new ideas for the development of various therapeutic drugs targeting Rac1.
Humans
;
rac1 GTP-Binding Protein/genetics*
;
Virus Replication
;
Glioma/pathology*
;
Actin Cytoskeleton/metabolism*
;
Animals
8.MAGED4B Promotes Glioma Progression via Inactivation of the TNF-α-induced Apoptotic Pathway by Down-regulating TRIM27 Expression.
Can LIU ; Jun LIU ; Juntang SHAO ; Cheng HUANG ; Xingliang DAI ; Yujun SHEN ; Weishu HOU ; Yuxian SHEN ; Yongqiang YU
Neuroscience Bulletin 2023;39(2):273-291
MAGED4B belongs to the melanoma-associated antigen family; originally found in melanoma, it is expressed in various types of cancer, and is especially enriched in glioblastoma. However, the functional role and molecular mechanisms of MAGED4B in glioma are still unclear. In this study, we found that the MAGED4B level was higher in glioma tissue than that in non-cancer tissue, and the level was positively correlated with glioma grade, tumor diameter, Ki-67 level, and patient age. The patients with higher levels had a worse prognosis than those with lower MAGED4B levels. In glioma cells, MAGED4B overexpression promoted proliferation, invasion, and migration, as well as decreasing apoptosis and the chemosensitivity to cisplatin and temozolomide. On the contrary, MAGED4B knockdown in glioma cells inhibited proliferation, invasion, and migration, as well as increasing apoptosis and the chemosensitivity to cisplatin and temozolomide. MAGED4B knockdown also inhibited the growth of gliomas implanted into the rat brain. The interaction between MAGED4B and tripartite motif-containing 27 (TRIM27) in glioma cells was detected by co-immunoprecipitation assay, which showed that MAGED4B was co-localized with TRIM27. In addition, MAGED4B overexpression down-regulated the TRIM27 protein level, and this was blocked by carbobenzoxyl-L-leucyl-L-leucyl-L-leucine (MG132), an inhibitor of the proteasome. On the contrary, MAGED4B knockdown up-regulated the TRIM27 level. Furthermore, MAGED4B overexpression increased TRIM27 ubiquitination in the presence of MG132. Accordingly, MAGED4B down-regulated the protein levels of genes downstream of ubiquitin-specific protease 7 (USP7) involved in the tumor necrosis factor-alpha (TNF-α)-induced apoptotic pathway. These findings indicate that MAGED4B promotes glioma growth via a TRIM27/USP7/receptor-interacting serine/threonine-protein kinase 1 (RIP1)-dependent TNF-α-induced apoptotic pathway, which suggests that MAGED4B is a potential target for glioma diagnosis and treatment.
Humans
;
Tumor Necrosis Factor-alpha
;
DNA-Binding Proteins/metabolism*
;
Ubiquitin-Specific Peptidase 7
;
Cisplatin
;
Temozolomide
;
Transcription Factors
;
Glioma
;
Cell Proliferation
;
Melanoma
;
Cell Line, Tumor
;
Apoptosis
;
Nuclear Proteins/genetics*
9.The Oncogenesis of Glial Cells in Diffuse Gliomas and Clinical Opportunities.
Qiyuan ZHUANG ; Hui YANG ; Ying MAO
Neuroscience Bulletin 2023;39(3):393-408
Glioma is the most common and lethal intrinsic primary tumor of the brain. Its controversial origins may contribute to its heterogeneity, creating challenges and difficulties in the development of therapies. Among the components constituting tumors, glioma stem cells are highly plastic subpopulations that are thought to be the site of tumor initiation. Neural stem cells/progenitor cells and oligodendrocyte progenitor cells are possible lineage groups populating the bulk of the tumor, in which gene mutations related to cell-cycle or metabolic enzymes dramatically affect this transformation. Novel approaches have revealed the tumor-promoting properties of distinct tumor cell states, glial, neural, and immune cell populations in the tumor microenvironment. Communication between tumor cells and other normal cells manipulate tumor progression and influence sensitivity to therapy. Here, we discuss the heterogeneity and relevant functions of tumor cell state, microglia, monocyte-derived macrophages, and neurons in glioma, highlighting their bilateral effects on tumors. Finally, we describe potential therapeutic approaches and targets beyond standard treatments.
Humans
;
Glioma/metabolism*
;
Neuroglia/metabolism*
;
Carcinogenesis/pathology*
;
Neural Stem Cells/metabolism*
;
Microglia/metabolism*
;
Brain Neoplasms/metabolism*
;
Tumor Microenvironment
10.RGS16 regulated by let-7c-5p promotes glioma progression by activating PI3K-AKT pathway.
Chaochao WANG ; Hao XUE ; Rongrong ZHAO ; Zhongzheng SUN ; Xiao GAO ; Yanhua QI ; Huizhi WANG ; Jianye XU ; Lin DENG ; Gang LI
Frontiers of Medicine 2023;17(1):143-155
Gliomas are the most common central nervous system tumours; they are highly aggressive and have a poor prognosis. RGS16 belongs to the regulator of G-protein signalling (RGS) protein family, which plays an important role in promoting various cancers, such as breast cancer, pancreatic cancer, and colorectal cancer. Moreover, previous studies confirmed that let-7c-5p, a well-known microRNA, can act as a tumour suppressor to regulate the progression of various tumours by inhibiting the expression of its target genes. However, whether RGS16 can promote the progression of glioma and whether it is regulated by miR let-7c-5p are still unknown. Here, we confirmed that RGS16 is upregulated in glioma tissues and that high expression of RGS16 is associated with poor survival. Ectopic deletion of RGS16 significantly suppressed glioma cell proliferation and migration both in vitro and in vivo. Moreover, RGS16 was validated as a direct target gene of miR let-7c-5p. The overexpression of miR let-7c-5p obviously downregulated the expression of RGS16, and knocking down miR let-7c-5p had the opposite effect. Thus, we suggest that the suppression of RGS16 by miR let-7c-5p can promote glioma progression and may serve as a potential prognostic biomarker and therapeutic target in glioma.
Humans
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
MicroRNAs/metabolism*
;
Glioma/genetics*
;
Genes, Tumor Suppressor
;
Cell Proliferation
;
Gene Expression Regulation, Neoplastic
;
Cell Line, Tumor

Result Analysis
Print
Save
E-mail