1.P4HA1 mediates YAP hydroxylation and accelerates collagen synthesis in temozolomide-resistant glioblastoma.
Xueru LI ; Gangfeng YU ; Xiao ZHONG ; Jiacheng ZHONG ; Xiangyu CHEN ; Qinglong CHEN ; Jinjiang XUE ; Xi YANG ; Xinchun ZHANG ; Yao LING ; Yun XIU ; Yaqi DENG ; Hongda LI ; Wei MO ; Yong ZHU ; Ting ZHANG ; Liangjun QIAO ; Song CHEN ; Fanghui LU
Chinese Medical Journal 2025;138(16):1991-2005
BACKGROUND:
Temozolomide (TMZ) resistance is a significant challenge in treating glioblastoma (GBM). Collagen remodeling has been shown to be a critical factor for therapy resistance in other cancers. This study aimed to investigate the mechanism of TMZ chemoresistance by GBM cells reprogramming collagens.
METHODS:
Key extracellular matrix components, including collagens, were examined in paired primary and recurrent GBM samples as well as in TMZ-treated spontaneous and grafted GBM murine models. Human GBM cell lines (U251, TS667) and mouse primary GBM cells were used for in vitro studies. RNA-sequencing analysis, chromatin immunoprecipitation, immunoprecipitation-mass spectrometry, and co-immunoprecipitation assays were conducted to explore the mechanisms involved in collagen accumulation. A series of in vitro and in vivo experiments were designed to assess the role of the collagen regulators prolyl 4-hydroxylase subunit alpha 1 (P4HA1) and yes-associated protein (YAP) in sensitizing GBM cells to TMZ.
RESULTS:
This study revealed that TMZ exposure significantly elevated collagen type I (COL I) expression in both GBM patients and murine models. Collagen accumulation sustained GBM cell survival under TMZ-induced stress, contributing to enhanced TMZ resistance. Mechanistically, P4HA1 directly binded to and hydroxylated YAP, preventing ubiquitination-mediated YAP degradation. Stabilized YAP robustly drove collagen type I alpha 1 ( COL1A1) transcription, leading to increased collagen deposition. Disruption of the P4HA1-YAP axis effectively reduced COL I deposition, sensitized GBM cells to TMZ, and significantly improved mouse survival.
CONCLUSION
P4HA1 maintained YAP-mediated COL1A1 transcription, leading to collagen accumulation and promoting chemoresistance in GBM.
Temozolomide
;
Humans
;
Glioblastoma/drug therapy*
;
Animals
;
Mice
;
Cell Line, Tumor
;
Drug Resistance, Neoplasm/genetics*
;
YAP-Signaling Proteins
;
Hydroxylation
;
Dacarbazine/pharmacology*
;
Adaptor Proteins, Signal Transducing/metabolism*
;
Transcription Factors/metabolism*
;
Collagen/biosynthesis*
;
Collagen Type I/metabolism*
;
Prolyl Hydroxylases/metabolism*
;
Antineoplastic Agents, Alkylating/therapeutic use*
2.Dysregulation of Iron Homeostasis Mediated by FTH Increases Ferroptosis Sensitivity in TP53-Mutant Glioblastoma.
Xuejie HUAN ; Jiangang LI ; Zhaobin CHU ; Hongliang ZHANG ; Lei CHENG ; Peng LUN ; Xixun DU ; Xi CHEN ; Qian JIAO ; Hong JIANG
Neuroscience Bulletin 2025;41(4):569-582
Iron metabolism is a critical factor in tumorigenesis and development. Although TP53 mutations are prevalent in glioblastoma (GBM), the mechanisms by which TP53 regulates iron metabolism remain elusive. We reveal an imbalance iron homeostasis in GBM via TCGA database analysis. TP53 mutations disrupted iron homeostasis in GBM, characterized by elevated total iron levels and reduced ferritin (FTH). The gain-of-function effect triggered by TP53 mutations upregulates itchy E3 ubiquitin-protein ligase (ITCH) protein expression in astrocytes, leading to FTH degradation and an increase in free iron levels. TP53-mut astrocytes were more tolerant to the high iron environment induced by exogenous ferric ammonium citrate (FAC), but the increase in intracellular free iron made them more sensitive to Erastin-induced ferroptosis. Interestingly, we found that Erastin combined with FAC treatment significantly increased ferroptosis. These findings provide new insights for drug development and therapeutic modalities for GBM patients with TP53 mutations from iron metabolism perspectives.
Ferroptosis/drug effects*
;
Humans
;
Iron/metabolism*
;
Glioblastoma/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Homeostasis/physiology*
;
Ferritins/metabolism*
;
Brain Neoplasms/genetics*
;
Mutation
;
Astrocytes/drug effects*
;
Cell Line, Tumor
;
Piperazines/pharmacology*
;
Quaternary Ammonium Compounds/pharmacology*
;
Ferric Compounds
3.FOXO1-miR-506 axis promotes chemosensitivity to temozolomide and suppresses invasiveness in glioblastoma through a feedback loop of FOXO1/miR-506/ETS1/FOXO1.
Chao CHEN ; Yu'e LIU ; Hongxiang WANG ; Xu ZHANG ; Yufeng SHI ; Juxiang CHEN
Journal of Zhejiang University. Science. B 2023;24(8):698-710
To explore the role of forkhead box protein O1 (FOXO1) in the progression of glioblastoma multiforme (GBM) and related drug resistance, we deciphered the roles of FOXO1 and miR-506 in proliferation, apoptosis, migration, invasion, autophagy, and temozolomide (TMZ) sensitivity in the U251 cell line using in vitro and in vivo experiments. Cell viability was tested by a cell counting kit-8 (CCK8) kit; migration and invasion were checked by the scratching assay; apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining and flow cytometry. The construction of plasmids and dual-luciferase reporter experiment were carried out to find the interaction site between FOXO1 and miR-506. Immunohistochemistry was done to check the protein level in tumors after the in vivo experiment. We found that the FOXO1-miR-506 axis suppresses GBM cell invasion and migration and promotes GBM chemosensitivity to TMZ, which was mediated by autophagy. FOXO1 upregulates miR-506 by binding to its promoter to enhance transcriptional activation. MiR-506 could downregulate E26 transformation-specific 1 (ETS1) expression by targeting its 3'-untranslated region (UTR). Interestingly, ETS1 promoted FOXO1 translocation from the nucleus to the cytosol and further suppressed the FOXO1-miR-506 axis in GBM cells. Consistently, both miR-506 inhibition and ETS1 overexpression could rescue FOXO1 overactivation-mediated TMZ chemosensitivity in mouse models. Our study demonstrated a negative feedback loop of FOXO1/miR-506/ETS1/FOXO1 in GBM in regulating invasiveness and chemosensitivity. Thus, the above axis might be a promising therapeutic target for GBM.
Animals
;
Mice
;
Brain Neoplasms/genetics*
;
Cell Line, Tumor
;
Cell Proliferation
;
Drug Resistance, Neoplasm
;
Feedback
;
Gene Expression Regulation, Neoplastic
;
Glioblastoma/metabolism*
;
MicroRNAs/metabolism*
;
Temozolomide/therapeutic use*
;
Humans
;
Forkhead Box Protein O1/metabolism*
4.Leucine-rich repeats containing 4 protein (LRRC4) in memory, psychoneurosis, and glioblastoma.
Chinese Medical Journal 2023;136(1):4-12
Leucine-rich repeats containing 4 ( LRRC4 , also named netrin-G ligand 2 [NGL-2]) is a member of the NetrinGs ligands (NGLs) family. As a gene with relatively high and specific expression in brain, it is a member of the leucine-rich repeat superfamily and has been proven to be a suppressor gene for gliomas, thus being involved in gliomagenesis. LRRC4 is the core of microRNA-dependent multi-phase regulatory loops that inhibit the proliferation and invasion of glioblastoma (GB) cells, including LRRC4/NGL2-activator protein 2 (AP2)-microRNA (miR) 182-LRRC4 and LRRC4-miR185-DNA methyltransferase 1 (DNMT1)-LRRC4/specific protein 1 (SP1)-DNMT1-LRRC4. In this review, we demonstrated LRRC4 as a new member of the partitioning-defective protein (PAR) polarity complex that promotes axon differentiation, mediates the formation and plasticity of synapses, and assists information input to the hippocampus and storage of memory. As an important synapse regulator, aberrant expression of LRRC4 has been detected in autism, spinal injury and GBs. LRRC4 is a candidate susceptibility gene for autism and a neuro-protective factor in spinal nerve damage. In GBs, LRRC4 is a novel inhibitor of autophagy, and an inhibitor of protein-protein interactions involving in temozolomide resistance, tumor immune microenvironment, and formation of circular RNA.
Humans
;
Cell Line, Tumor
;
Glioblastoma/metabolism*
;
Leucine
;
Leucine-Rich Repeat Proteins/genetics*
;
MicroRNAs
;
Nerve Tissue Proteins/genetics*
;
Tumor Microenvironment
5.Effect of Morus alba extract sanggenon C on growth and proliferation of glioblastoma cells.
Wen-Han TANG ; Zhi-Ning ZHANG ; Hua-Rui CAI ; Wei SUN ; He YANG ; Er-Hu ZHAO ; Hong-Juan CUI
China Journal of Chinese Materia Medica 2023;48(1):211-219
Glioblastoma is the most common primary cranial malignancy, and chemotherapy remains an important tool for its treatment. Sanggenon C(San C), a class of natural flavonoids extracted from Morus plants, is a potential antitumor herbal monomer. In this study, the effect of San C on the growth and proliferation of glioblastoma cells was examined by methyl thiazolyl tetrazolium(MTT) assay and 5-bromodeoxyuridinc(BrdU) labeling assay. The effect of San C on the tumor cell cycle was examined by flow cytometry, and the effect of San C on clone formation and self-renewal ability of tumor cells was examined by soft agar assay. Western blot and bioinformatics analysis were used to investigate the mechanism of the antitumor activity of San C. In the presence of San C, the MTT assay showed that San C significantly inhibited the growth and proliferation of tumor cells in a dose and time-dependent manner. BrdU labeling assay showed that San C significantly attenuated the DNA replication activity in the nucleus of tumor cells. Flow cytometry confirmed that San C blocked the cell cycle of tumor cells in G_0/G_1 phase. The soft agar clone formation assay revealed that San C significantly attenuated the clone formation and self-renewal ability of tumor cells. The gene set enrichment analysis(GSEA) implied that San C inhibited the tumor cell division cycle by affecting the myelocytomatosis viral oncogene(MYC) signaling pathway. Western blot assay revealed that San C inhibited the expression of cyclin through the regulation of the MYC signaling pathway by lysine demethylase 4B(KDM4B), which ultimately inhibited the growth and proliferation of glioblastoma cells and self-renewal. In conclusion, San C exhibits the potential antitumor activity by targeting the KDM4B-MYC axis to inhibit glioblastoma cell growth, proliferation, and self-renewal.
Humans
;
Glioblastoma/genetics*
;
Bromodeoxyuridine/therapeutic use*
;
Signal Transduction
;
Proto-Oncogene Proteins c-myc/metabolism*
;
Agar
;
Cell Proliferation
;
Cell Line, Tumor
;
Apoptosis
;
Jumonji Domain-Containing Histone Demethylases/metabolism*
6.The action mechanism of glioblastoma cell-derived exosome: a review.
Na LI ; Li LUO ; Yating YANG ; Zhaomei LIU ; Xiaoyan QIU ; Mingyu WANG ; Wei WANG ; Xiong XIAO
Chinese Journal of Biotechnology 2023;39(4):1477-1501
Patients with glioblastoma (GBM) generally have a bad prognosis and short overall survival after being treated with surgery, chemotherapy or radiotherapy due to the histological heterogeneity, strong invasive ability and rapid postoperative recurrence of GBM. The components of GBM cell-derived exosome (GBM-exo) can regulate the proliferation and migration of GBM cell via cytokines, miRNAs, DNA molecules and proteins, promote the angiogenesis via angiogenic proteins and non-coding RNAs, mediate tumor immune evasion by targeting immune checkpoints with regulatory factors, proteins and drugs, and reduce drug resistance of GBM cells through non-coding RNAs. GBM-exo is expected to be an important target for the personalized treatment of GBM and a marker for diagnosis and prognosis of this kind of disease. This review summarizes the preparation methods, biological characteristics, functions and molecular mechanisms of GBM-exo on cell proliferation, angiogenesis, immune evasion and drug resistance of GBM to facilitate developing new strategies for the diagnosis and treatment of GBM.
Humans
;
Glioblastoma/genetics*
;
Exosomes/metabolism*
;
MicroRNAs/metabolism*
;
Prognosis
;
Cell Proliferation
;
Brain Neoplasms/genetics*
;
Cell Line, Tumor
7.Specific Regulation of m6A by SRSF7 Promotes the Progression of Glioblastoma.
Yixian CUN ; Sanqi AN ; Haiqing ZHENG ; Jing LAN ; Wenfang CHEN ; Wanjun LUO ; Chengguo YAO ; Xincheng LI ; Xiang HUANG ; Xiang SUN ; Zehong WU ; Yameng HU ; Ziwen LI ; Shuxia ZHANG ; Geyan WU ; Meisongzhu YANG ; Miaoling TANG ; Ruyuan YU ; Xinyi LIAO ; Guicheng GAO ; Wei ZHAO ; Jinkai WANG ; Jun LI
Genomics, Proteomics & Bioinformatics 2023;21(4):707-728
Serine/arginine-rich splicing factor 7 (SRSF7), a known splicing factor, has been revealed to play oncogenic roles in multiple cancers. However, the mechanisms underlying its oncogenic roles have not been well addressed. Here, based on N6-methyladenosine (m6A) co-methylation network analysis across diverse cell lines, we find that the gene expression of SRSF7 is positively correlated with glioblastoma (GBM) cell-specific m6A methylation. We then indicate that SRSF7 is a novel m6A regulator, which specifically facilitates the m6A methylation near its binding sites on the mRNAs involved in cell proliferation and migration, through recruiting the methyltransferase complex. Moreover, SRSF7 promotes the proliferation and migration of GBM cells largely dependent on the presence of the m6A methyltransferase. The two m6A sites on the mRNA for PDZ-binding kinase (PBK) are regulated by SRSF7 and partially mediate the effects of SRSF7 in GBM cells through recognition by insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2). Together, our discovery reveals a novel role of SRSF7 in regulating m6A and validates the presence and functional importance of temporal- and spatial-specific regulation of m6A mediated by RNA-binding proteins (RBPs).
Humans
;
Cell Line, Tumor
;
Cell Proliferation
;
Gene Expression Regulation, Neoplastic
;
Glioblastoma/genetics*
;
Methyltransferases/metabolism*
;
RNA Splicing Factors/metabolism*
;
RNA, Messenger/genetics*
;
RNA-Binding Proteins/metabolism*
;
Serine-Arginine Splicing Factors/metabolism*
;
RNA Methylation/genetics*
8.Single-cell transcriptome analysis of multigrade glioma heterogeneity and immune microenvironment revealed potential prognostic biomarkers.
Jie LIU ; Kailong XU ; Lixin MA ; Yang WANG
Chinese Journal of Biotechnology 2022;38(10):3790-3808
Glioma, the most common intrinsic tumor of the central nervous system, is characterized by its high incidence and poor prognosis. The aim of this study was to identify differentially expressed genes (DEGs) between glioblastoma multiforme (GBM) and low-grade glioma (LGG) to explore prognostic factors of different grades of gliomas. Single-cell transcriptome sequencing data of gliomas were collected from the NCBI Gene Expression Omnibus (GEO), which included a total of 29 097 cell samples from three datasets. For the analysis of human gliomas of different grades, 21 071 cells were obtained by filtering, and 70 genes were screened from differentially expressed genes by gene ontology (GO) analysis, Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis, from which the gene DLL3 was focused by reviewing the literature. The TCGA-based gene expression profiling interactive analysis (GEPIA) database was used to explore the survival curves of genes in LGG and GBM, and the gene expression profiling interactive analysis and tumor immune estimation resource (TIMER) database was used to study the expression of key genes in gliomas of different grades, predicting biomarkers that were closely related to immunotherapy. The cBioPortal database was used to explore the relationship between DLL3 expression and 25 immune checkpoints. Gene set enrichment analysis (GSEA) further identified pathways associated with central genes. Finally, the efficacy of biomarkers in prognosis and prediction was validated in the Chinese glioma genome atlas (CGGA). These results demonstrated that prognostic genes are associated with tumor proliferation and progression. Analysis of biological information and survival suggested that these genes might serve as a promising prognostic biomarker and as new targets for selecting therapeutic strategies.
Humans
;
Biomarkers
;
Brain Neoplasms/pathology*
;
Gene Expression Profiling/methods*
;
Glioblastoma/pathology*
;
Glioma/pathology*
;
Intracellular Signaling Peptides and Proteins
;
Membrane Proteins/genetics*
;
Prognosis
;
Transcriptome
;
Tumor Microenvironment/genetics*
;
Biomarkers, Tumor
9.Intratumor heterogeneity, microenvironment, and mechanisms of drug resistance in glioma recurrence and evolution.
Zhaoshi BAO ; Yongzhi WANG ; Qiangwei WANG ; Shengyu FANG ; Xia SHAN ; Jiguang WANG ; Tao JIANG
Frontiers of Medicine 2021;15(4):551-561
Glioma is the most common lethal tumor of the human brain. The median survival of patients with primary World Health Organization grade IV glioma is only 14.6 months. The World Health Organization classification of tumors of the central nervous system categorized gliomas into lower-grade gliomas and glioblastomas. Unlike primary glioblastoma that usually develop de novo in the elderly, secondary glioblastoma enriched with an isocitrate dehydrogenase mutant typically progresses from lower-grade glioma within 5-10 years from the time of diagnosis. Based on various evolutional trajectories brought on by clonal and subclonal alterations, the evolution patterns of glioma vary according to different theories. Some important features distinguish the normal brain from other tissues, e.g., the composition of the microenvironment around the tumor cells, the presence of the blood-brain barrier, and others. The underlying mechanism of glioma recurrence and evolution patterns of glioma are different from those of other types of cancer. Several studies correlated tumor recurrence with tumor heterogeneity and the immune microenvironment. However, the detailed reasons for the progression and recurrence of glioma remain controversial. In this review, we introduce the different mechanisms involved in glioma progression, including tumor heterogeneity, the tumor microenvironment and drug resistance, and their pre-clinical implements in clinical trials. This review aimed to provide new insights into further clinical strategies for the treatment of patients with recurrent and secondary glioma.
Aged
;
Brain Neoplasms/genetics*
;
Drug Resistance
;
Glioblastoma
;
Glioma/genetics*
;
Humans
;
Mutation
;
Neoplasm Recurrence, Local/drug therapy*
;
Tumor Microenvironment
10.Overexpression of lncRNA MEG3 inhibits proliferation and invasion of glioblastoma U251 cells
Qizhi LUO ; Fan ZHANG ; Wei LI ; Fang WANG ; Lixiang WU ; Baisheng HUANG
Journal of Southern Medical University 2021;41(1):141-145
OBJECTIVE:
To investigate the effects of overexpression of long noncoding RNA (lncRNA) MEG3 on the proliferation and invasion of glioblastoma U251 cells by suppressing the expression of hypoxia inducible factor 1
METHODS:
The expression of lncRNA MEG3 and HIF1
RESULTS:
The expression of MEG3 was significantly lower and HIF1
CONCLUSIONS
MEG3 overexpression inhibits the proliferation and invasion of U251 cells through suppressing the expression of HIF1
Apoptosis
;
Cell Line, Tumor
;
Cell Movement/genetics*
;
Cell Proliferation/genetics*
;
Gene Expression Regulation, Neoplastic
;
Glioblastoma/genetics*
;
Humans
;
MicroRNAs
;
RNA, Long Noncoding/genetics*

Result Analysis
Print
Save
E-mail