1
|
Wen J, Wu D, Le Y, Yin Z, Chen M, Shen Y, Wu X, Liu K, Luo K, Shu Z, Shu Q, Ouyang D. Engineered nanovesicles targeting SERPINE1 overcome temozolomide resistance in glioblastoma. Cell Signal 2025; 132:111763. [PMID: 40139622 DOI: 10.1016/j.cellsig.2025.111763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/15/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor with limited treatment options due to its resistance to temozolomide (TMZ). This study explores a novel therapeutic approach using engineered cell membrane nanovesicles loaded with SERPINE1 inhibitors to combat TMZ resistance. High-throughput sequencing identified pivotal genes associated with resistance, while the nanovesicles demonstrated excellent stability and the ability to cross the blood-brain barrier. Functional assays revealed significant suppression of GBM cell viability, migration, and invasion, accompanied by reduced expression of SERPINE1 and VEGF, suggesting inhibition of angiogenesis and tumor progression. These findings highlight the potential of SERPINE1-targeted nanovesicles as an innovative and effective strategy for overcoming TMZ resistance in GBM.
Collapse
Affiliation(s)
- Jianping Wen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China.
| | - Dongxu Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yi Le
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zonghua Yin
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Minglong Chen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yulong Shen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Xia Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kebo Liu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kun Luo
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zhicheng Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Qingxia Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Dongsheng Ouyang
- The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
2
|
Tan B, Chen T, Song P, Lin F, He S, Yin X. lncRNA BBOX1-AS1 regulates the miR-382-5p/CBX3 Signalling pathway to affect the proliferation and apoptosis of glioblastoma cells. Int Immunopharmacol 2025; 157:114790. [PMID: 40339498 DOI: 10.1016/j.intimp.2025.114790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
OBJECTIVE This study aims to investigate the role of long noncoding RNA (lncRNA) BBOX1-AS1 in regulating the miR-382-5p/CBX3 axis and its impact on glioblastoma cell proliferation and apoptosis. METHODS U-87 MG glioblastoma cells were divided into the following groups: control, si-NC, si-BBOX1-AS1, si-BBOX1-AS1 + inhibitor NC, si-BBOX1-AS1 + miR-382-5p inhibitor, miR-NC, miR-382-5p mimic, miR-382-5p mimic+pc-NC, and miR-382-5p mimic+pc-CBX3. The expression levels of lncRNAs BBOX1-AS1, miR-382-5p, and CBX3 were detected via qRT-PCR. Cell proliferation was assessed via MTT assays. Apoptosis rates and caspase 3 activity were measured via flow cytometry and caspase 3 activity assay kits. Western blotting was used to evaluate CBX3 protein expression. Dual-luciferase reporter assays were conducted to verify the relationship between the lncRNA BBOX1-AS1 and miR-382-5p. A similar assay was performed for miR-382-5p and CBX3. RESULTS Compared to normal human astrocytes (HA1800), glioblastoma cells (T98G, HS 683, U251, and U87 MG) exhibited higher levels of lncRNA BBOX1-AS1 and CBX3, along with lower levels of miR-382-5p (P < 0.05). Compared with the si-NC cells, the U-87 MG cells in the si-BBOX1-AS1 group presented reduced lncRNA BBOX1-AS1 expression, proliferation ability, and CBX3 protein expression but increased miR-382-5p levels, apoptosis rates, and Caspase 3 activity (P < 0.05). Compared with the si-BBOX1-AS1 + inhibitor NC group, the si-BBOX1-AS1 + miR-382-5p inhibitor group presented decreased miR-382-5p levels, apoptosis rates, and Caspase 3 activity but increased CBX3 protein expression and proliferation ability (P < 0.05). Compared with the control, the miR-382-5p mimic reduced CBX3 protein expression and proliferation ability while increasing the apoptosis rate and caspase 3 activity (P < 0.05). However, overexpression of CBX3 (miR-382-5p mimic+pc-CBX3) reversed the effects of the miR-382-5p mimic (P < 0.05). Dual-luciferase assays confirmed the relationships between lncRNA BBOX1-AS1 and miR-382-5p and between miR-382-5p and CBX3 (P < 0.05). CONCLUSION Knockdown of lncRNA BBOX1-AS1 inhibits U-87 MG cell proliferation and promotes apoptosis by upregulating miR-382-5p and downregulating CBX3 expression.
Collapse
Affiliation(s)
- Bo Tan
- Neurosurgery, Guangyuan Central Hospital, No.16 jinghangzi, Guangyaun, 628000, Sichuan, China
| | - Tao Chen
- Neurosurgery, Guangyuan Central Hospital, No.16 jinghangzi, Guangyaun, 628000, Sichuan, China
| | - Peng Song
- Neurosurgery, Guangyuan Central Hospital, No.16 jinghangzi, Guangyaun, 628000, Sichuan, China
| | - Feng Lin
- Neurosurgery, Guangyuan Central Hospital, No.16 jinghangzi, Guangyaun, 628000, Sichuan, China
| | - Shuangyin He
- Neurosurgery, Guangyuan Central Hospital, No.16 jinghangzi, Guangyaun, 628000, Sichuan, China
| | - Xiaohong Yin
- Neurosurgery, Guangyuan Central Hospital, No.16 jinghangzi, Guangyaun, 628000, Sichuan, China.
| |
Collapse
|
3
|
Wang S, Sun Z, Wang C, Zhang A, Zhang C, Hou S, Lin N, Li Q. The JAK1/STAT3 pathway mediates the effects of SERPINH1 on glioma EMT. Int Immunopharmacol 2025; 157:114731. [PMID: 40334628 DOI: 10.1016/j.intimp.2025.114731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
Glioma is marked by swift cell proliferation, extensive invasion and poor outcomes. Serine protease inhibitor H1 (SERPINH1) encoding heat shock protein 47, a collagen-specific molecular chaperone, plays a role in a number of cancers. However, its definite role in glioma remains unclear. The aim of the present study was to investigate the role of SERPINH1 in glioma progression, especially its impact on cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT). The glioma cell lines LN229, T98, U251 and U87MG were transfected with lentivirus for stable knockdown or overexpression of SERPINH1. Assays assessing cell proliferation, migration and invasion were conducted to investigate the role of SERPINH1 in these processes. Bioinformatic analysis was conducted using The Cancer Genome Atlas and the Chinese Glioma Genome Atlas databases to identify potential molecular pathways associated with SERPINH1. Western blotting (WB) was employed to examine the expression of significant proteins in the JAK1/STAT3 signaling pathway and EMT markers. Nude mice were used for in vivo experiments to evaluate tumor growth and changes related to EMT. Overexpression of SERPINH1 notably increased glioma cell proliferation, migration and invasion, whereas knockdown suppressed these activities. Bioinformatic analyses revealed that SERPINH1 is closely associated with the JAK1/STAT3 signaling pathway and EMT-related genes. WB results confirmed that SERPINH1 regulates the activation of JAK1/STAT3 and influences the levels of EMT markers such as N- and E-cadherin. The JAK1/STAT3 agonist RO8191 partially rescued glioma cell behavior in the SERPINH1 knockdown group, while the inhibitor STATTIC partially weakened the enhanced effects in the SERPINH1 overexpression group. In vivo, SERPINH1 overexpression accelerated tumor growth and EMT progression, while knockdown resulted in a reduction in tumor size and the expression of EMT markers. SERPINH1 is essential for glioma progression, enhancing cell proliferation, migration, invasion and EMT by activating the JAK1/STAT3 signaling pathway. These results indicate that targeting SERPINH1 could provide a promising new approach for glioma therapy.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Zhiming Sun
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Chao Wang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Antian Zhang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Chao Zhang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Shiqiang Hou
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Ning Lin
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China.
| | - Qun Li
- Health Examination Center, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China.
| |
Collapse
|
4
|
Joma N, Kagelmacher M, Zhang I, Herrmann A, Dernedde J, Haag R, Maysinger D. Charged dendrimers reduce glioblastoma viability by modulating lysosomal activity and HMGB1-RAGE interaction. Biochem Pharmacol 2025; 238:116969. [PMID: 40348093 DOI: 10.1016/j.bcp.2025.116969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
Dendrimers and dendrimer-based self-assembly systems have emerged as promising nanocarriers for a variety of applications, including anti-cancer therapies, modulation of the tumor microenvironment, and imaging. Here, we explored the therapeutic potential of two charged dendrimers, dendritic polyglycerol sulfate (dPGS) and dendritic polyglycerol amine (dPGA), in the context of glioblastoma multiforme (GBM). Docosahexaenoic acid (DHA) has shown potential in GBM. We therefore examined dPGS and dPGA effects alone and in combination with DHA. Using 2D cell models and 3D tumoroids, we showed that DHA with dPGA reduced tumor integrity and cell viability. dPGS reduced oxidative stress, whereas dPGA reduced lysosomal acidification, contributing to cellular dysfunction. Both dendrimers influence the interaction between high mobility group box 1 (HMGB1) and the receptor for advanced glycation end products (RAGE). The surfaces of the HMGB1-RAGE complex provide binding sites for interactions of charged molecules like dPGS and dPGA, suggesting the contribution of these interactions to cytotoxicity. In summary, our findings show that combining DHA with charged dendrimers (dPGS and dPGA) enhances GBM cytotoxicity through several mechanisms, involving lysosomal alkalinization, lipid peroxidation and modulation of the HMGB1-RAGE complex.
Collapse
Affiliation(s)
- Natali Joma
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Marten Kagelmacher
- Clinical Chemistry and Pathobiochemistry, Charité University Medicine Berlin, Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Andreas Herrmann
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.
| | - Jens Dernedde
- Clinical Chemistry and Pathobiochemistry, Charité University Medicine Berlin, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.
| |
Collapse
|
5
|
Liu L, Wuyun T, Sun X, Zhang Y, Cha G, Zhao L. Therapeutic efficacy of TMTP1-modified EVs in overcoming bone metastasis and immune resistance in PIK3CA mutant NSCLC. Cell Death Dis 2025; 16:367. [PMID: 40328748 PMCID: PMC12055990 DOI: 10.1038/s41419-025-07685-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
Non-small cell lung cancer (NSCLC) with PIK3CA mutations demonstrates significant challenges in treatment due to enhanced bone metastasis and immune checkpoint resistance. This study investigates the efficacy of tumor-targeting peptide 1-modified cancer stem cell-derived extracellular vesicles (TMTP1-TSRP-EVs) in reshaping the tumor microenvironment and reversing immune checkpoint resistance in NSCLC. By integrating TMTP1-TSRP into EVs, we aim to specifically deliver therapeutic agents to NSCLC cells, focusing on inhibiting the PI3K/Akt/mTOR pathway, a crucial driver of oncogenic activity and immune evasion in PIK3CA-mutated cells. Our comprehensive in vitro and in vivo analyses show that TMTP1-TSRP-EVs significantly inhibit tumor growth, reduce PD-L1 expression, and enhance CD8+ T cell infiltration, effectively reversing the immune-suppressive microenvironment. Moreover, the in vivo models confirm that our approach not only suppresses bone metastases but also overcomes primary resistance to immune checkpoint inhibitors by modulating the expression of key immunological markers. These findings suggest that targeted delivery of TMTP1-TSRP-EVs could provide a novel therapeutic strategy for treating PIK3CA-mutant NSCLC, offering significant improvements over traditional therapies by directly targeting the molecular pathogenesis of tumor resistance and metastasis. Molecular Mechanisms Reshaping the TME to Halt PI3K-Mutant Bone Metastasis of NSCLC and Overcoming Primary ICI Resistance. (Created by BioRender).
Collapse
Affiliation(s)
- Liwen Liu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tanghesi Wuyun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Sun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Zhang
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Geqi Cha
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ling Zhao
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
6
|
Guan Y, Luan Y, Zhao S, Li M, Girolamo F, Palmer JD, Guan Q. Single-cell RNA sequencing for characterizing the immune communication and iron metabolism roles in CD31 + glioma cells. Transl Cancer Res 2025; 14:2421-2439. [PMID: 40386270 PMCID: PMC12079608 DOI: 10.21037/tcr-2025-377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/08/2025] [Indexed: 05/20/2025]
Abstract
Background Gliomas are aggressive brain tumors marked by complex cellular interactions and significant immune cell infiltration. This study investigated the role of CD31+ immune cells, specifically macrophages and T cells, in the glioma microenvironment through single-cell RNA sequencing (scRNA-seq). Methods We employed the CellChat framework to map cell-cell communication pathways and used Monocle3 for pseudotime trajectory analysis to characterize the signaling and developmental progressions within CD31+ cells. Pathways such as osteopontin (SPP1) and major histocompatibility complex class II (MHC-II) were analyzed in terms of their role in immune regulation, and we examined the expression of ferritin, an iron-binding protein, to assess its potential function in modulating CD31+ cell activity. Results Our findings highlight the expression of key pathways, including SPP1 and MHC-II, influencing immune regulation. Ferritin was found to be highly expressed in CD31+ cells, suggesting a dual role in iron metabolism and immune modulation within the glioma microenvironment. Conclusions This study clarified the distinct roles of CD31+ immune cells in glioma progression and identified ferritin as a potential therapeutic target for modulating immune responses in gliomas. These findings may offer new directions in glioma research and the development of immunotherapy, which can aid in improving treatment outcomes.
Collapse
Affiliation(s)
- Yiming Guan
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Yu Luan
- Clinical Laboratory Center, The First People’s Hospital of Shenyang (Shenyang Brain Hospital), Shenyang Medical College, Shenyang, China
| | - Shanshan Zhao
- Clinical Laboratory Center, The First People’s Hospital of Shenyang (Shenyang Brain Hospital), Shenyang Medical College, Shenyang, China
| | - Meiyan Li
- Tuberculosis Laboratory, Shenyang Tenth People’s Hospital (Shenyang Chest Hospital), Shenyang, China
| | - Francesco Girolamo
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Bari, Italy
| | - Joshua D. Palmer
- Department of Radiation Oncology, Ohio State University, Columbus, OH, USA
| | - Qi Guan
- Clinical Laboratory Center, The First People’s Hospital of Shenyang (Shenyang Brain Hospital), Shenyang Medical College, Shenyang, China
| |
Collapse
|
7
|
Fu W, Liang Q, Ma Y, Lei S, Li R, Zheng X, Chen L, Chen J, Cai X, Dai X, Duan H, He W, Ren J. Fn14-targeting, NIR-II responsive nanomaterials for enhanced radiotherapy against glioblastomas. NANOSCALE ADVANCES 2025; 7:2634-2647. [PMID: 40109505 PMCID: PMC11915457 DOI: 10.1039/d4na00788c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/01/2025] [Indexed: 03/22/2025]
Abstract
Radiotherapy is a common treatment option for patients with glioblastoma multiforme. However, tumor heterogeneity causes varying responses to radiation among different tumor subpopulations. Cancer cells that endure radiotherapy exhibit radioresistance, resulting in the ineffectiveness of radiation therapy and eventual tumor relapse. In this study, we discovered that the fibroblast growth factor-inducible 14 (Fn14)-positive tumor cells were enriched in tumor residual foci after radiation, ultimately leading to treatment failure. Fn14-expressing glioma cells survived ionizing radiation through preferential activation of DNA damage checkpoint response. We have thus engineered an Fn14-targeting and NIR-II responsive plasmonic gold nanosystem named Fn14-AuNPs, which can precisely internalize into Fn14-overexpressed glioma cells and have an excellent BBB-crossing capability. As gold nanoparticles, by inhibition of DNA repair processes and induction of G2/M cells cycle arrest, Fn14-AuNPs nanoparticles improved the radiosensitivity of tumor cells. Meanwhile, Fn14-AuNPs induced localized heat under NIR-II photoirradiation, thus impeding RT-induced DNA damage checkpoint response. This versatile nanosensitizer, combined with NIR-II laser photoirradiation, can eradicate radioresistant subpopulations of glioblastoma and improve the therapeutic effect of radiotherapy. This finding presents an effective radiosensitization strategy by targeting radioresistant subpopulations, which can efficiently overcome the constraints imposed in clinical radiotherapy and offer a hopeful avenue to enhance the treatment effectivity of radiotherapy in glioblastoma.
Collapse
Affiliation(s)
- Wei Fu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Yuxi Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Shiqiong Lei
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
| | - Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Lian Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Jiayuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xing Cai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xiaofang Dai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637457 Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore 636921 Singapore
| | - Wenshan He
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
- Shenzhen Huazhong University of Science and Technology Research Institute Shenzhen 518057 China
| |
Collapse
|
8
|
Sandhbor P, John G, Bhat S, Goda JS. Immune response recalibration using immune therapy and biomimetic nano-therapy against high-grade gliomas and brain metastases. Asian J Pharm Sci 2025; 20:101021. [PMID: 40224727 PMCID: PMC11987628 DOI: 10.1016/j.ajps.2025.101021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/07/2024] [Accepted: 10/03/2024] [Indexed: 04/15/2025] Open
Abstract
Although with aggressive standards of care like surgical resection, chemotherapy, and radiation, high-grade gliomas (HGGs) and brain metastases (BM) treatment has remained challenging for more than two decades. However, technological advances in this field and immunotherapeutic strategies have revolutionized the treatment of HGGs and BM. Immunotherapies like immune checkpoint inhibitors, CAR-T targeting, oncolytic virus-based therapy, bispecific antibody treatment, and vaccination approaches, etc., are emerging as promising avenues offering new hope in refining patient's survival benefits. However, selective trafficking across the blood-brain barrier (BBB), immunosuppressive tumor microenvironment (TME), metabolic alteration, and tumor heterogeneity limit the therapeutic efficacy of immunotherapy for HGGs and BM. Furthermore, to address this concern, the NanoBioTechnology-based bioinspired delivery system has been gaining tremendous attention in recent years. With technological advances such as Trojan horse targeting and infusing/camouflaging nanoparticles surface with biological molecules/cells like immunocytes, erythrocytes, platelets, glioma cell lysate and/or integrating these strategies to get hybrid membrane for homotypic recognition. These biomimetic nanotherapy offers advantages over conventional nanoparticles, focusing on greater target specificity, increased circulation stability, higher active loading capacity, BBB permeability (inherent inflammatory chemotaxis of neutrophils), decreased immunogenicity, efficient metabolism-based combinatorial effects, and prevention of tumor recurrence by induction of immunological memory, etc. provide new age of improved immunotherapies outcomes against HGGs and BM. In this review, we emphasize on neuro-immunotherapy and the versatility of these biomimetic nano-delivery strategies for precise targeting of hard-to-treat and most lethal HGGs and BM. Moreover, the challenges impeding the clinical translatability of these approaches were addressed to unmet medical needs of brain cancers.
Collapse
Affiliation(s)
- Puja Sandhbor
- Institute for NanoBioTechnology, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore 21218, USA
| | - Geofrey John
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Kharghar 410210, India
- Homi Bhabha National Institute, Anushakti Nagar 400094, India
| | - Sakshi Bhat
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Kharghar 410210, India
- Homi Bhabha National Institute, Anushakti Nagar 400094, India
| | - Jayant S. Goda
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Kharghar 410210, India
- Homi Bhabha National Institute, Anushakti Nagar 400094, India
| |
Collapse
|
9
|
Wen J, Wu X, Shu Z, Wu D, Yin Z, Chen M, Luo K, Liu K, Shen Y, Le Y, Shu Q. Clusterin-mediated polarization of M2 macrophages: a mechanism of temozolomide resistance in glioblastoma stem cells. Stem Cell Res Ther 2025; 16:146. [PMID: 40128761 PMCID: PMC11934612 DOI: 10.1186/s13287-025-04247-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
Glioblastoma remains one of the most lethal malignancies, largely due to its resistance to standard chemotherapy such as temozolomide. This study investigates a novel resistance mechanism involving glioblastoma stem cells (GSCs) and the polarization of M2-type macrophages, mediated by the extracellular vesicle (EV)-based transfer of Clusterin. Using 6-week-old male CD34+ humanized huHSC-(M-NSG) mice (NM-NSG-017) and glioblastoma cell lines (T98G and U251), we demonstrated that GSC-derived EVs enriched with Clusterin induce M2 macrophage polarization, thereby enhancing temozolomide resistance in glioblastoma cells. Single-cell and transcriptome sequencing revealed close interactions between GSCs and M2 macrophages, highlighting Clusterin as a key mediator. Our findings indicate that Clusterin-rich EVs from GSCs drive glioblastoma cell proliferation and resistance to temozolomide by modulating macrophage phenotypes. Targeting this pathway could potentially reverse resistance mechanisms, offering a promising therapeutic approach for glioblastoma. This study not only sheds light on a critical pathway underpinning glioblastoma resistance but also lays the groundwork for developing therapies targeting the tumor microenvironment. Our results suggest a paradigm shift in understanding glioblastoma resistance, emphasizing the therapeutic potential of disrupting EV-mediated communication in the tumor microenvironment.
Collapse
Affiliation(s)
- Jianping Wen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China.
| | - Xia Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Zhicheng Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Dongxu Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Zonghua Yin
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Minglong Chen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Kun Luo
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Kebo Liu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Yulong Shen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Yi Le
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China
| | - Qingxia Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, No. 144, Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, China.
| |
Collapse
|
10
|
Manu DR, Bǎlaşa R, Pruteanu LL, Curean V, Barbu-Tudoran L, Şerban GM, Chinezu R, Bǎlaşa A. Identification of distinct profiles of glioblastoma through the immunocapture of extracellular vesicles from patient plasma. PLoS One 2025; 20:e0315890. [PMID: 40106404 PMCID: PMC11922215 DOI: 10.1371/journal.pone.0315890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/03/2024] [Indexed: 03/22/2025] Open
Abstract
Glioblastoma (GBM), a primary brain tumor, exhibits intratumoral heterogeneity and dynamic spatial-temporal changes. GBM-derived extracellular vesicles (EVs), reflecting tumor characteristics, present potential as liquid-biopsy markers for early diagnosis and monitoring. This study aims to evaluate molecular signatures of plasma-derived EVs from GBM patients using a conventional flow cytometer. EVs have been isolated from glioma patients and healthy controls (HCs) plasma using density gradient ultracentrifugation (DGU). EVs were evaluated by bead-based multiplex analysis in a conventional flow cytometer. Principal component analysis (PCA), hierarchical clustering, and correlation analysis provided comprehensive insights into EV characteristics. EVs successfully isolated were visualized in transmission and scanning electron microscopy (STEM). Bead-based multiplex analysis in flow cytometer detected the level of 37 EV surface markers, including tumor-related, cancer stem cell, endothelial cell, and immune cell- specific antigens. PCA identified the EV surface markers that are most significant for differentiating the subjects, and hierarchical clustering revealed four distinct clusters based on EV surface marker levels. EV molecular signature demonstrated considerable heterogeneity across patient clusters. The presence of CD29 emerged not only as a defining factor for a cluster of patients, but also served as a marker to differentiate patients from HCs.
Collapse
Affiliation(s)
- Doina Ramona Manu
- Center for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, Targu Mures, Romania,
| | - Rodica Bǎlaşa
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, Targu Mures, Romania
- 1st Neurology Clinic, Emergency Clinical County Hospital of Targu Mures, Targu Mures, Romania
| | - Lavinia-Lorena Pruteanu
- Department of Chemistry and Biology, North University Center at Baia Mare, Technical University of Cluj-Napoca, Baia Mare, Romania
- Research Center for Functional Genomics, Biomedicine, and Translational Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Victor Curean
- Doctoral School, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Barbu-Tudoran
- Department of Molecular Biology and Biotechnology, Electron Microscopy Laboratory, Biology and Geology Faculty, Babes-Bolyai University, Cluj-Napoca, Romania
- Electron Microscopy Integrated Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| | - Georgiana-Mihaela Şerban
- Doctoral School, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, Targu Mures, Romania
| | - Rareş Chinezu
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, Romania
- Neurosurgery Clinic, Emergency Clinical County Hospital of Targu Mures, Targu Mures, Romania
| | - Adrian Bǎlaşa
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, Romania
- Neurosurgery Clinic, Emergency Clinical County Hospital of Targu Mures, Targu Mures, Romania
| |
Collapse
|
11
|
Shah DD, Chorawala MR, Raghani NR, Patel R, Fareed M, Kashid VA, Prajapati BG. Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med Oncol 2025; 42:117. [PMID: 40102282 DOI: 10.1007/s12032-025-02641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Tumor microenvironment (TME) denotes the non-cancerous cells and components presented in the tumor, including molecules produced and released by them. Interactions between cancer cells, immune cells, stromal cells, and the extracellular matrix within the TME create a dynamic ecosystem that can either promote or hinder tumor growth and spread. The TME plays a pivotal role in either promoting or inhibiting tumor growth and dissemination, making it a critical factor to consider in the development of effective cancer therapies. Understanding the intricate interplay within the TME is crucial for devising effective cancer therapies. Combination therapies involving inhibitors of immune checkpoint blockade (ICB), and/or chemotherapy now offer new approaches for cancer therapy. However, it remains uncertain how to best utilize these strategies in the context of the complex tumor microenvironment. Oncogene-driven changes in tumor cell metabolism can impact the TME to limit immune responses and present barriers to cancer therapy. Cellular and acellular components in tumor microenvironment can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Components in the TME can reprogram tumor behavior and influence responses to treatments, facilitating immune evasion, nutrient deprivation, and therapeutic resistance. Moreover, the TME can influence angiogenesis, promoting the formation of blood vessels that sustain tumor growth. Notably, the TME facilitates immune evasion, establishes a nutrient-deprived milieu, and induces therapeutic resistance, hindering treatment efficacy. A paradigm shift from a cancer-centric model to a TME-centric one has revolutionized cancer research and treatment. However, effectively targeting specific cells or pathways within the TME remains a challenge, as the complexity of the TME poses hurdles in designing precise and effective therapies. This review highlights challenges in targeting the tumor microenvironment to achieve therapeutic efficacy; explore new approaches and technologies to better decipher the tumor microenvironment; and discuss strategies to intervene in the tumor microenvironment and maximize therapeutic benefits.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Rajanikant Patel
- Department of Product Development, Granules Pharmaceuticals Inc., 3701 Concorde Parkway, Chantilly, VA, 20151, USA
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 13713, Riyadh, Saudi Arabia
| | - Vivekanand A Kashid
- MABD Institute of Pharmaceutical Education and Research, Babhulgaon, Yeola, Nashik, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
12
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
13
|
Fang X, Jia H, Pan S, Liu Q, Wang Q, Feng Y, Ding W, Luo T. Matrix Stiffness Regulates Interleukin-10 Secretion in Human Microglia (HMC3) via YAP-Mediated Mechanotransduction. Cell Biochem Funct 2025; 43:e70061. [PMID: 40011226 DOI: 10.1002/cbf.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Microglia, as resident immune cells in the brain, adhere to the extracellular matrix and typically exhibit anti-inflammatory polarization under normal physiological conditions. Despite their pivotal roles, the regulatory effects of extracellular matrix properties on microglial function and the associated molecular mechanisms remain inadequately understood. Here, we elucidate how matrix stiffness modulates interleukin-10 (IL-10) secretion in human microglia (HMC3) via yes-associated protein (YAP)-mediated mechanotransduction. Using soft collagen Ⅰ-coated hydrogels, we observed a substantial reduction in IL-10 secretion, accompanied by a decrease in the expression and nuclear localization of YAP compared to cells adhered to glass substrates. With increasing hydrogel substrate stiffness, the expression and nuclear localization of YAP were enhanced, leading to an elevated secretion of IL-10. Subsequently, to further investigate the relationship between YAP and IL-10, we performed YAP depletion experiments, which revealed that nuclear exclusion of YAP suppressed IL-10 secretion. Interestingly, overexpression of YAP in microglia did not markedly affect IL-10 levels. We seeded YAP-knockdown microglia onto hydrogels of varying stiffness, and no significant differences were observed in IL-10 secretion. Our findings suggested that cytoskeletal polymerization was crucial for the regulation of IL-10 secretion mediated by YAP. Given the crucial role of IL-10 in the tumor microenvironment, we further found shYAP-microglia attenuated the pro-proliferative effect of microglia on gliomas. Besides, when YAP was silenced, actin of human microglia decreased, and their contractility was weakened. In summary, this study identifies YAP as a pivotal molecule in controlling cytokine secretion and sensing matrix stiffness in microglia. These insights offer potential therapeutic avenues for glioma treatment by targeting YAP-mediated pathways in microglial cells.
Collapse
Affiliation(s)
- Xue Fang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Haiying Jia
- Chinese People's Liberation Army Strategic Support Force Special Medical Center, Beijing, China
| | - Shaoshan Pan
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Qian Liu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Qian Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Ye Feng
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Weiping Ding
- School of Information Science and Technology, University of Science and Technology of China, Hefei, China
| | - Tianzhi Luo
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
- The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
| |
Collapse
|
14
|
Wang W, Li J, He Q, Liu C, Wang S, Zheng Z, Zhang B, Mou S, Sun W, Zhao J. Integrated Analysis to Reveal Heterogeneity of Tumor-Associated Neutrophils in Glioma. Cancer Med 2025; 14:e70745. [PMID: 40052358 PMCID: PMC11886415 DOI: 10.1002/cam4.70745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/20/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Glioma, characterized by its cellular and molecular heterogeneity, presents formidable challenges in treatment strategy and prognostic assessment. The tumor microenvironment (TME) profoundly influences tumor behavior and treatment response, with tumor-associated neutrophils (TANs) playing a complex but understudied role. This study aimed to investigate the heterogeneity and role of TANs in glioma and to develop a prognostic model. METHODS Analysis of scRNA-seq data identified cellular subpopulations and differentially expressed neutrophil-related genes (DE-NRGs). Bulk RNA-seq was obtained from four independent datasets. Molecular subtypes of glioma samples were determined by consensus clustering. WGCNA was conducted to elucidate the association between gene modules and subtypes. We developed a risk score model. Expression of selected genes was confirmed using immunohistochemistry (IHC). In vitro experiments were also performed for functional verification, including CCK8, EdU, Transwell, and TUNEL assays. RESULTS A total of 108 DE-NRGs for TANs were identified based on scRNA-seq data. Two molecular subtypes were characterized, showing significant differences in prognosis and clinical features. Immune-related analyses demonstrated varied immunological characteristics between subtypes. The risk score model was constructed with 7 genes, including AEBP1, CAVIN1, DCTD, DEPP1, DUSP6, FKBP9, and UGCG. It showed significant prognostic value and was validated across three external datasets. The mutation landscape highlighted higher IDH mutation prevalence in low-risk groups. Drug sensitivity analysis indicated TMZ resistance in high-risk groups. In vitro experiments showed that UGCG could promote glioma cell proliferation, migration, and invasion, while decreasing apoptosis. CONCLUSION This study explored the heterogeneity of TANs and developed a prognostic model, providing insights for prognostic prediction and guiding personalized treatment strategies in glioma. Declaration of Generative AI in Scientific Writing: The authors declare nonuse of generative AI and AI-assisted technologies in the writing process.
Collapse
Affiliation(s)
- Wen Wang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Junsheng Li
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qiheng He
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Chenglong Liu
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Siyu Wang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Zhiyao Zheng
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Bojian Zhang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Siqi Mou
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Wei Sun
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jizong Zhao
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| |
Collapse
|
15
|
Yue Y, Ren Y, Lu C, Jiang N, Wang S, Fu J, Kong M, Zhang G. The research progress on meningeal metastasis in solid tumors. Discov Oncol 2025; 16:254. [PMID: 40019647 PMCID: PMC11871263 DOI: 10.1007/s12672-025-01950-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/08/2024] [Indexed: 03/01/2025] Open
Abstract
Meningeal metastasis (MM), particularly Leptomeningeal metastases (LM), represents the advanced stage of solid tumors and poses a significant threat to patients' lives. Moreover, it imposes a substantial burden on society. LM represents the ultimate and most fatal stage of solid tumors, inflicting devastating consequences on patients and imposing a substantial burden on society. The incidence of LM continues to rise annually, emphasizing the urgent need for early recognition and treatment initiation in individuals with LM to significantly extend overall patient survival. Despite rapid advancements in current LM detection and treatment methods, the diagnosis of LM remains constrained by several limitations such as low diagnostic efficiency, the therapeutic outcomes remain suboptimal. Furthermore, there is currently no universally recognized industry standard for LM treatment, further underscoring its status as an unresolved challenge in tumor management. Additionally, progress towards elucidating the mechanisms underlying MM has stagnated. Therefore, this review aims to comprehensively summarize recent research advances pertaining to MM in solid tumors by elucidating its underlying mechanisms, exploring diagnostic and prognostic biomarkers while addressing existing research challenges.
Collapse
Affiliation(s)
- Yi Yue
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chunya Lu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Nan Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Sihui Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Junkai Fu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Mengrui Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
16
|
Zheng Y, Shi J. EFNB1 drives glioma progression and shapes the immune microenvironment: a potential prognostic biomarker. Discov Oncol 2025; 16:249. [PMID: 40014231 PMCID: PMC11868007 DOI: 10.1007/s12672-025-01867-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/03/2025] [Indexed: 02/28/2025] Open
Abstract
Gliomas, a heterogeneous group of tumors affecting the brain and spinal cord, present a significant clinical challenge. Ephrin B1 (EFNB1) has been implicated in various malignancies. However, its role in gliomas remained poorly understood. Hence, this study aimed to elucidate the connection between EFNB1 and the progression of glioma. A retrospective RNA-seq analysis was conducted by utilizing the data from glioma patients in the TCGA and CGGA databases. Kaplan-Meier survival analysis and multivariate regression models were employed to evaluate the prognostic significance of EFNB1. RT-PCR was used to quantify EFNB1 expression in glioma tissues and cell lines. Meanwhile, in vitro assays were carried out to assess its functional roles in glioma cells. Our findings demonstrated that EFNB1 expression was significantly elevated in gliomas and other cancers. Moreover, high EFNB1 expression was closely correlated with advanced clinical stages and poor prognosis. Notably, multivariate analysis identified EFNB1 as an independent prognostic factor for overall survival. KEGG pathway analysis suggested that EFNB1 was involved in critical biological processes, including the cell cycle, protein processing in the endoplasmic reticulum, Epstein-Barr virus infection, and Salmonella infection. Furthermore, EFNB1 expression was associated with immune cell infiltration, particularly Th2 cells, macrophages, and plasmacytoid dendritic cells. In glioma cells, EFNB1 expression was markedly increased. Consequently, functional experiments demonstrated that EFNB1 knockdown inhibited glioma cell proliferation, invasion, and migration. These results highlighted EFNB1 as a novel independent prognostic biomarker and suggest its potential role in shaping the immunological microenvironment of gliomas.
Collapse
Affiliation(s)
- Yungui Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, China
| | - Jiasong Shi
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, China.
| |
Collapse
|
17
|
Liu H, Tan S, Zhao Z, Tang X, Li Z, Qi J. METTL3/YTDHF1 Stabilizes MTCH2 mRNA to Regulate Ferroptosis in Glioma Cells. FRONT BIOSCI-LANDMRK 2025; 30:25718. [PMID: 40018930 DOI: 10.31083/fbl25718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Gliomas are aggressive brain tumors known for their poor prognosis and resistance to standard treatment options. Ferroptosis is an iron-dependent form of regulated cell death that has emerged as a promising target for cancer treatment. This study examined how the methyltransferase-like 3/YTH domain family protein 1 (METTL3/YTHDF1) axis influences ferroptosis and glioma progression by stabilizing mitochondrial carrier homolog 2 (MTCH2) messenger RNA (mRNA). METHODS MTCH2 expression in glioma tissues and cell lines was evaluated through quantitative real-time polymerase chain reaction (PCR) and western blot analyses. To assess the effects of MTCH2 knockdown and overexpression on glioma cell functions, we performed a series of functional assays, including cell viability, colony formation, and measurements of lipid reactive oxygen species (lipid ROS) and malondialdehyde (MDA) levels. Additionally, we conducted RNA immunoprecipitation (RIP) and RNA stability assays to explore the underlying mechanisms governing the interaction between METTL3, YTHDF1, and the stability of MTCH2 mRNA. RESULTS MTCH2 was significantly upregulated in glioma tissues and cell lines. Silencing of MTCH2 resulted in decreased glioma cell proliferation and induced ferroptosis, as evidenced by increased lipid peroxidation and ROS accumulation. Conversely, overexpression of MTCH2 enhanced glioma cell survival and reduced ferroptosis. METTL3-mediated N6-methyladenosine (m6A) modification enhanced MTCH2 mRNA stability by enabling YTHDF1 to bind and protect the modified mRNA from degradation. CONCLUSION The METTL3/YTHDF1/MTCH2 axis plays a critical role in glioma progression by inhibiting ferroptosis and promoting tumor cell survival. Targeting this pathway may provide a new and effective treatment strategy for glioma patients.
Collapse
Affiliation(s)
- Hongjun Liu
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Shasha Tan
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Zhenyu Zhao
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Xiaoping Tang
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Zhou Li
- Department of Neurosurgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Jian Qi
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| |
Collapse
|
18
|
Rosito M, Maqbool J, Reccagni A, Mangano M, D'Andrea T, Rinaldi A, Peruzzi G, Silvestri B, Rosa A, Trettel F, D'Alessandro G, Catalano M, Fucile S, Limatola C. Ketogenic diet induces an inflammatory reactive astrocytes phenotype reducing glioma growth. Cell Mol Life Sci 2025; 82:73. [PMID: 39921723 PMCID: PMC11807044 DOI: 10.1007/s00018-025-05600-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/23/2024] [Accepted: 01/16/2025] [Indexed: 02/10/2025]
Abstract
The use of a ketogenic diet (KD) in glioma is currently tested as an adjuvant treatment in standard chemotherapy regimens. The metabolic shift induced by the KD leads to the generation of ketone bodies that can influence glioma cells and the surrounding microenvironment, but the mechanisms have not yet been fully elucidated. Here, we investigated the potential involvement of glial cells as mediators of the KD-induced effects on tumor growth and survival rate in glioma-bearing mice. Specifically, we describe that exposing glioma-bearing mice to a KD or to β-hydroxybutyrate (β-HB), one of the main KD metabolic products, reduced glioma growth in vivo, induced a pro-inflammatory phenotype in astrocytes and increased functional glutamate transporters. Moreover, we described increased intracellular basal Ca2+ levels in GL261 glioma cells treated with β-HB or co-cultured with astrocytes. These data suggest that pro-inflammatory astrocytes triggered by β-HB can be beneficial in counteracting glioma proliferation and neuronal excitotoxicity, thus protecting brain parenchyma.
Collapse
Affiliation(s)
- Maria Rosito
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy.
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy.
| | - Javeria Maqbool
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Alice Reccagni
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Micol Mangano
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | | | - Arianna Rinaldi
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Beatrice Silvestri
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Alessandro Rosa
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Flavia Trettel
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Sergio Fucile
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, IS, Italy.
- Department of Physiology and Pharmacology, Laboratory Affiliated to Institute Pasteur Italia, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
19
|
Yu S, Wu J, Jing Y, Lin P, Lang L, Xiong Y, Chen W, Liu W, Sun C, Lu Y. Research trends in glioma chemoradiotherapy resistance: a bibliometric analysis (2003-2023). Front Oncol 2025; 15:1539937. [PMID: 39990688 PMCID: PMC11842341 DOI: 10.3389/fonc.2025.1539937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
Background Glioma is the most aggressive primary malignant tumor of the central nervous system, characterized by high recurrence rates and resistance to chemoradiotherapy, making therapeutic resistance a major challenge in neuro-oncology. Recent research emphasizes the role of the tumor microenvironment (TME) and immune modulation in glioma progression and resistance. Despite these advances, a comprehensive bibliometric analysis of research trends in glioma chemoradiotherapy resistance over the past two decades is lacking. This study aims to systematically evaluate the research landscape, identify emerging hotspots, and provide guidance for future investigations. Methods Articles on glioma chemoradiotherapy resistance published between 2003 and 2023 were retrieved from the Web of Science Core Collection, resulting in 4,528 publications. Bibliometric tools, including VOSviewer, CiteSpace, and R packages such as bibliometrix and ggplot2, were used to analyze co-authorship networks, keyword evolution, and citation bursts to identify collaboration patterns, thematic developments, and influential contributions. Results Publication output increased significantly between 2013 and 2022, peaking at 650 articles in 2022. Over 1,000 institutions from 88 countries contributed to this research. The United States, Switzerland, and Germany showed the highest citation impact, while China led in publication volume but demonstrated relatively lower citation influence. The research focus has shifted from traditional topics such as the "MGMT gene" to emerging areas including the "tumor microenvironment," "immune infiltration," and "nanoparticles." The androgen receptor was identified as a promising but underexplored therapeutic target. Conclusions Research on glioma chemoradiotherapy resistance has seen substantial growth, with increasing emphasis on immune modulation, the tumor microenvironment, and novel therapeutic targets such as the androgen receptor. This study represents the first comprehensive bibliometric analysis of this field, providing a detailed overview of research trends and potential directions for future studies. The findings highlight the need for strengthened international collaboration and multidisciplinary approaches to address the challenges of therapeutic resistance in glioma.
Collapse
Affiliation(s)
- Shishi Yu
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinya Wu
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuan Jing
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Lin
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Lang Lang
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yifan Xiong
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Wangzhong Chen
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhua Liu
- Clinical Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changpeng Sun
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuntao Lu
- Nanfang hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Liao J, Duan Y, Xu X, Liu Y, Zhan C, Xiao G. Circadian rhythm related genes signature in glioma for drug resistance prediction: a comprehensive analysis integrating transcriptomics and machine learning. Discov Oncol 2025; 16:119. [PMID: 39909964 PMCID: PMC11799505 DOI: 10.1007/s12672-025-01863-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/03/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Gliomas, 24% of all primary brain tumors, have diverse histology and poor survival rates, with about 70% recurring due to acquired or de novo resistance. Insomnia in patients is correlated strongly with circadian rhythm disruptions. The correlation between circadian rhythm disorders and drug resistance of some tumors has been proved. However, the precise mechanism underlying the relationship between glioma and circadian rhythm disorders has not been elucidated. METHODS Circadian rhythm-related genes (CRRGs) were identified using the least absolute shrinkage and selection operator (LASSO) regression, and stochastic gradient descent (SGD) was performed to form a circadian rhythm-related score (CRRS) model. The studies of immune cell infiltration, genetic variations, differential gene expression pattern, and single cell analysis were performed for exploring the mechanisms of chemotherapy resistance in glioma. The relationship between CRRGs and chemosensitivity was also confirmed by IC 50 (half maximal inhibitory concentration) analysis. RESULT Signatures of 16 CRRGs were screened out and identified. Based on the CRRS model, an optimal comprehensive nomogram was created, exhibiting a favorable potential for predicting drug resistance in samples. Immune infiltration, cell-cell communication, and single cell analysis all indicated that high CRRS group was closely related to innate immune cells. IC50 analysis showed that CRRG knockdown enhanced the chemosensitivity of glioma. CONCLUSION A significant correlation between CRRGs, drug resistance of glioma, and innate immune cells was found, which might hold a significant role in the drug resistance of glioma.
Collapse
Affiliation(s)
- Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingxing Duan
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Xiangwang Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaxue Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chaohong Zhan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
21
|
Stojanovic NM, Mitić M, Ilić J, Radić M, Radisavljević M, Baralić M, Krstić M. Natural Source of Drugs Targeting Central Nervous System Tumors-Focus on NAD(P)H Oxidoreductase 1 (NQO1) Activity. Brain Sci 2025; 15:132. [PMID: 40002465 PMCID: PMC11853159 DOI: 10.3390/brainsci15020132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Central nervous system (CNS) tumors involve a large and diverse group of malignancies that arise from various cell types within the brain tissue. Although there are advances in treatments, CNS tumors still remain challenging, due to their complex biology and the delicate nature of the surrounding tissue. NAD(P)H O=oxidoreductase 1 (NQO1) is an enzyme that plays a critical role in the detoxification of quinones, protecting cells from oxidative stress. In CNS tumors this enzyme is often overexpressed, which contributes to the resistance of tumor cells to chemotherapy by enhancing their antioxidant defenses. NQO1 influences the progression of CNS tumors by affecting downstream signaling pathways, such as those involving the transcription factor SNAIL, as well as others that are associated with tumor behavior. Plants represent a valuable source of numerous constituents with different chemical structures known to affect different molecular signaling pathways associated with different pathologies.
Collapse
Affiliation(s)
- Nikola M. Stojanovic
- Department of Physiology, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
| | - Milica Mitić
- Center for Pathology, University Clinical Centre Niš, 18000 Niš, Serbia; (M.M.); (M.K.)
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (J.I.); (M.R.); (M.R.)
| | - Jovan Ilić
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (J.I.); (M.R.); (M.R.)
- Department for Neurosurgery, University Clinical Centre Niš, 18000 Niš, Serbia
| | - Milica Radić
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (J.I.); (M.R.); (M.R.)
- Department for Radiation Oncology, University Clinical Centre Niš, 18000 Niš, Serbia
| | - Miša Radisavljević
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (J.I.); (M.R.); (M.R.)
- Department for Neurosurgery, University Clinical Centre Niš, 18000 Niš, Serbia
| | - Marko Baralić
- Faculty of Medicine, University of Belgrade, 11080 Belgrade, Serbia;
- Department of Nephrology, University Clinical Centre of Serbia, 11000 Belgrade, Serbia
| | - Miljan Krstić
- Center for Pathology, University Clinical Centre Niš, 18000 Niš, Serbia; (M.M.); (M.K.)
- Department of Pathology, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
| |
Collapse
|
22
|
Luo K, Zhuang K, Wu H, Chen Y, Liu Y, Yang F, Wang Z. PLIN1 suppresses glioma progression through regulating lipid metabolism. Cell Death Dis 2025; 16:48. [PMID: 39870645 PMCID: PMC11772837 DOI: 10.1038/s41419-025-07347-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 12/29/2024] [Accepted: 01/10/2025] [Indexed: 01/29/2025]
Abstract
Glioma is a common and destructive brain tumor, which is highly heterogeneous with poor prognosis. Developing diagnostic and prognostic markers to identify and treat glioma early would significantly improve the therapeutic outcomes. Here, we conducted RNA next-generation sequencing with 33 glioma samples and 15 normal brain samples. We found Perilipin 1 (PLIN1) downregulated in glioma and correlated with poorer outcome. Subsequent experiments revealed that up regulation of PLIN1 led to repressed cell growth and invasion in glioma. Moreover, overexpression of PLIN1 increased lipid accumulation in glioma cells, with increasing expression of lipid biosynthesis related genes and decreasing expression of lipolysis related genes. Mechanically, we revealed that the PI3K/AKT axis could regulate PLIN1 levels in glioma, that inhibition of the activity of PI3K/AKT axis could increase PLIN1 levels in glioma. In conclusion, the dysregulation PI3K/AKT axis led to PLIN1 downregulation and the following tumor proliferation, invasion and lipid metabolism reprogramming in glioma.
Collapse
Affiliation(s)
- Kui Luo
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kai Zhuang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Wu
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanbing Chen
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Liu
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fan Yang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhifei Wang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
23
|
Saadh MJ, Ghnim ZS, Mahdi MS, Chandra M, Ballal S, Bareja L, Chaudhary K, Sharma RSK, Gupta S, Taher WM, Alwan M, Jawad MJ, Hamad AK. Decoding the Role of Kinesin Superfamily Proteins in Glioma Progression. J Mol Neurosci 2025; 75:10. [PMID: 39847238 DOI: 10.1007/s12031-025-02308-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/04/2025] [Indexed: 01/24/2025]
Abstract
Glioma is a highly aggressive and invasive brain tumor with limited treatment options, highlighting the need for novel therapeutic approaches. Kinesin superfamily proteins (KIFs) are a diverse group of motor proteins that play essential roles in cellular processes such as mitosis, intracellular transport, and signal transduction, all of which are crucial for tumorigenesis. This review focuses on the multifaceted role of KIFs in glioma, examining their clinical relevance, contribution to tumor progression, and potential as therapeutic targets. We discuss how KIFs influence key aspects of glioma biology, including cell proliferation, invasion, migration, and metastasis. Furthermore, we explore the regulation of the cell cycle and critical signaling pathways associated with glioma, such as PI3K-Akt, Wnt/β-catenin, and Hedgehog signaling by KIFs. The review also addresses the emerging interplay between KIFs and non-coding RNAs, including circular RNAs (circRNAs) and microRNAs (miRNAs), in glioma progression. Finally, we examine current therapeutic strategies targeting KIFs, including immunotherapy, chemotherapy, and small-molecule inhibitors, and their potential to improve treatment outcomes for glioma patients. By synthesizing these insights, this review underscores the significance of KIFs in glioma pathogenesis and their promise as novel therapeutic targets in the fight against glioma.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | | | - Muktesh Chandra
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - R S K Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Sofia Gupta
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
24
|
Horta M, Soares P, Leite Pereira C, Lima RT. Emerging Approaches in Glioblastoma Treatment: Modulating the Extracellular Matrix Through Nanotechnology. Pharmaceutics 2025; 17:142. [PMID: 40006509 PMCID: PMC11859630 DOI: 10.3390/pharmaceutics17020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Glioblastoma's (GB) complex tumor microenvironment (TME) promotes its progression and resistance to therapy. A critical component of TME is the extracellular matrix (ECM), which plays a pivotal role in promoting the tumor's invasive behavior and aggressiveness. Nanotechnology holds significant promise for GB treatment, with the potential to address challenges posed by both the blood-brain barrier and the GB ECM. By enabling targeted delivery of therapeutic and diagnostic agents, nanotechnology offers the prospect of improving treatment efficacy and diagnostic accuracy at the tumor site. This review provides a comprehensive exploration of GB, including its epidemiology, classification, and current treatment strategies, alongside the intricacies of its TME. It highlights nanotechnology-based strategies, focusing on nanoparticle formulations such as liposomes, polymeric nanoparticles, and gold nanoparticles, which have shown promise in GB therapy. Furthermore, it explores how different emerging nanotechnology strategies modulate the ECM to overcome the challenges posed by its high density, which restricts drug distribution within GB tumors. By emphasizing the intersection of nanotechnology and GB ECM, this review underscores an innovative approach to advancing GB treatment. It addresses the limitations of current therapies, identifies new research avenues, and emphasizes the potential of nanotechnology to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Horta
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Paula Soares
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Catarina Leite Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Raquel T. Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
25
|
Huang X, Yu G, Jiang X, Shen F, Wang D, Wu S, Mi Y. ITGB4/GNB5 axis promotes M2 macrophage reprogramming in NSCLC metastasis. Int Immunopharmacol 2025; 144:113564. [PMID: 39577216 DOI: 10.1016/j.intimp.2024.113564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/21/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE Metastasis of non-small cell lung cancer (NSCLC) is a leading cause of high mortality. In recent years, the role of M2 macrophages in promoting tumor metastasis within the tumor microenvironment has garnered increasing attention. This study aims to investigate the role and potential mechanisms of the ITGB4/GNB5 axis in regulating M2 macrophage reprogramming and influencing NSCLC metastasis. METHODS This study first used single-cell sequencing technology to reveal the diverse subpopulation structure of NSCLC tumor tissues. Data analysis then identified the correlation between M2 macrophages and the malignant phenotype of NSCLC. Flow cytometry and immunohistochemistry were used to detect changes in M2 macrophages in NSCLC tissues. The impact of the ITGB4/GNB5 axis on M2 macrophage function was assessed through RNA sequencing and proteomic analysis. Finally, in vitro cell experiments and in vivo mouse models were used to validate the function and regulatory mechanisms of this axis. RESULTS Our study found diverse cellular subpopulations in NSCLC tumor tissues, with M2 macrophages closely associated with the malignant phenotype of NSCLC. We identified ITGB4 as a characteristic gene of NSCLC and predicted GNB5 as an interacting gene through database analysis. Activation of the ITGB4/GNB5 axis was shown to enhance M2 macrophage polarization, promoting their accumulation in the tumor microenvironment. This change further facilitated NSCLC invasion and metastasis by modulating related cytokines and signaling pathways. Animal experiments demonstrated that inhibition of the ITGB4/GNB5 axis significantly reduced tumor growth and metastasis. CONCLUSION The ITGB4/GNB5 axis reshapes the TME by promoting M2 macrophage polarization and functional enhancement, thereby facilitating tumor invasion and metastasis in NSCLC. This research provides new insights into the molecular mechanisms of NSCLC and offers potential molecular targets for future targeted therapies.
Collapse
Affiliation(s)
- Xiaofeng Huang
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Guiping Yu
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Xuewei Jiang
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Fei Shen
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Dengshu Wang
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Song Wu
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China.
| | - Yedong Mi
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China.
| |
Collapse
|
26
|
Tang H, Yang X, Li G, Peng K, Sun Y, Jiang L, Huang Y. Development and experimental validation of dephosphorylation-related biomarkers to assess prognosis and immunotherapeutic response in gliomas. Front Immunol 2025; 15:1488894. [PMID: 39830513 PMCID: PMC11739095 DOI: 10.3389/fimmu.2024.1488894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Background Gliomas are common aggressive brain tumors with poor prognosis. Dephosphorylation-related biomarkers are in a void in gliomas. This study aims to construct a validated prognostic risk model for dephosphorylation, which will provide new directions for clinical treatment, prognostic assessment, and temozolomide (TMZ) resistance in glioma patients. Methods Screening dephosphorylation-related genes (DRGs) and transcriptome expression data from The Cancer Genome Atlas (TCGA), Molecular signatures database (MSigDB) and constructing risk scoring models. Kaplan-Meier (K-M), nomogram and ROC curve were used to assess the predictive efficacy of the model. Gene set enrichment analysis (GSEA), immune cell infiltration, immunotherapy response and chemotherapeutic drug sensitivity analysis were performed in this study. The correlation between chemotherapeutic drugs and the half maximal inhibitory concentration (IC50) values of 12 DRGs was analyzed. Cell division cycle 25A (CDC25A) and TMZ were screened and verified by experiments. Quantitative Real-Time PCR (qRT-PCR) detection of mRNA expression of 12 genes in human normal glial cells and two glioma cell lines. Transfection techniques overexpressed and knocked down CDC25A. qRT-PCR and Western Blot (WB) were used to detect the mRNA and protein expression levels of CDC25A. Subsequently, verify the effect of CDC25A on TMZ resistance in glioma cells. Results The model established in this study was able to accurately predict the prognosis of glioma patients. Besides, there were significant differences in GSEA, immune cell infiltration, immunotherapeutic response and chemotherapeutic drug sensitivity analysis between glioma patients in the high and low risk groups. The results of CCK8 experiments showed that overexpression of CDC25A increased the susceptibility of U251 and LN229 cells to TMZ, and knockdown of CDC25A attenuated the susceptibility of U251 and LN229 cells to TMZ.
Collapse
Affiliation(s)
- Hui Tang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Guoqian Li
- Department of Pharmacy, Chengdu Chengnan Jinhua Hospital, Chengdu, China
| | - Ke Peng
- Department of Pharmacy, Renshou County Traditional Chinese Medicine Hospital, Meshan, China
| | - Yang Sun
- Department of Pharmaceutical Analysis, Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, China
| | - Longyang Jiang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
27
|
Wu Y, Li N, Shang J, Jiang J, Liu X. Identification of cancer-associated fibroblast subtypes and prognostic model development in breast cancer: role of the RUNX1/SDC1 axis in promoting invasion and metastasis. Cell Biol Toxicol 2025; 41:21. [PMID: 39753834 PMCID: PMC11698906 DOI: 10.1007/s10565-024-09950-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/20/2024] [Indexed: 01/06/2025]
Abstract
In this study, we identified cancer-associated fibroblast (CAF) molecular subtypes and developed a CAF-based prognostic model for breast cancer (BRCA). The heterogeneity of cancer-associated fibroblasts (CAFs) and their significant involvement in the advancement of BRCA were discovered employing single-cell RNA sequencing. Notably, we discovered that the RUNX1/SDC1 axis enhances BRCA cell invasion and metastasis. RUNX1 transcriptionally upregulates SDC1, which facilitates extracellular matrix remodeling and promotes tumor cell migration. This finding highlights the vital contribution of CAFs to the tumor microenvironment and provides new potential targets for therapeutic intervention. The predictive model showcased remarkable precision in anticipating patient outcomes and could guide personalized treatment strategies.
Collapse
Affiliation(s)
- Yunhao Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Pancreatic and Thyroid Ward, Shenyang, 110004, P. R. China
| | - Nu Li
- Department of Breast surgery, The First Hospital of China Medical University, Shenyang, 110004, P.R. China
| | - Jin Shang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, P. R. China
| | - Jiazi Jiang
- Department of Emergency, The First Hospital of China Medical University, No.155 Nanjing Road, Heping District, Shenyang, 110001, Liaoning Province, P. R. China.
| | - Xiaoliang Liu
- Department of Emergency, The First Hospital of China Medical University, No.155 Nanjing Road, Heping District, Shenyang, 110001, Liaoning Province, P. R. China.
| |
Collapse
|
28
|
Wang W, Li J, He Q, Liu C, Zheng Z, Zhang B, Mou S, Sun W, Zhao J. Crosstalk between CD180-overexpression macrophages and glioma cells worsens patient survival through malignant phenotype promotion and immunosuppressive regulation. Mol Med 2024; 30:264. [PMID: 39707188 DOI: 10.1186/s10020-024-01029-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Understanding the molecular mechanisms in immunosuppressive regulation is crucial for improving immunotherapeutic strategies. Macrophages, the major immune cells in tumor microenvironment (TME), play a dual role in tumor progression. CD180, primarily expressed in macrophages, remains unclear and requires further investigation. METHODS RNA-seq data were obtained to analyze CD180 expression in gliomas and assess its prognostic value. The comprehensive immune infiltration analysis was performed. Single-cell RNA-seq (scRNA-seq) data were used to examine CD180 expression distribution at the cellular level. CD180-overexpression macrophages were co-cultured with three glioma cell lines. The effects on glioma cell behavior were evaluated through qRT-PCR, Western blot, CCK-8 assay, EdU assay, Transwell assay, TUNEL assay, and flow cytometry. Differentially expressed genes (DEGs) and their potential biological functions were analyzed between different CD180 expression groups. Consensus clustering was used to identify CD180-related glioma subtypes. RESULTS CD180 was significantly upregulated in glioma samples and associated with poor prognosis. High CD180 expression was correlated with increased immune cell infiltration, particularly macrophages, and elevated levels of immune checkpoints. Analysis of scRNA-seq data revealed the predominant expression of CD180 in macrophages within the glioma TME. In vitro experiments demonstrated that CD180-overexpression macrophages promoted glioma cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), while decreasing apoptosis. Mutations in TP53 and PTEN were significantly more prevalent in the high CD180 expression group. We identified nine chemotherapeutic agents that were more effective in glioma patients with high CD180 expression. Additionally, two CD180-related glioma subtypes with distinct prognosis were identified. CONCLUSIONS This study confirmed the prognostic role of CD180 in glioma and its involvement in immunosuppressive regulation and malignant phenotype promotion. Therefore, CD180 was considered as a promising target for immunotherapeutic strategies in glioma treatment.
Collapse
Affiliation(s)
- Wen Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhiyao Zheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Bojian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Siqi Mou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
29
|
Luo Y, Gadd ME, Qie Y, Otamendi-Lopez A, Sanchez-Garavito JE, Brooks MM, Ulloa Navas MJ, Hundal T, Li S, Jones VK, Lou Y, Patel T, Dronca R, Kharfan-Dabaja MA, Dong H, Quinones-Hinojosa A, Qin H. Solid cancer-directed CAR T cell therapy that attacks both tumor and immunosuppressive cells via targeting PD-L1. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200891. [PMID: 39498357 PMCID: PMC11532918 DOI: 10.1016/j.omton.2024.200891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has encountered limited success in solid tumors. The lack of dependable antigens and the immunosuppressive tumor microenvironment (TME) are major challenges. Within the TME, tumor cells along with immunosuppressive cells employ an immune-evasion mechanism that upregulates programmed death ligand 1 (PD-L1) to deactivate effector T cells; this makes PD-L1 a reliable, universal target for solid tumors. We developed a novel PD-L1 CAR (MC9999) using our humanized anti-PD-L1 monoclonal antibody, designed to simultaneously target tumor and immunosuppressive cells. The antigen-specific antitumor effects of MC9999 CAR T cells were observed consistently across four solid tumor models: breast cancer, lung cancer, melanoma, and glioblastoma multiforme (GBM). Notably, intravenous administration of MC9999 CAR T cells eradicated intracranially established LN229 GBM tumors, suggesting penetration of the blood-brain barrier. The proof-of-concept data demonstrate the cytolytic effect of MC9999 CAR T cells against immunosuppressive cells, including microglia HMC3 cells and M2 macrophages. Furthermore, MC9999 CAR T cells elicited cytotoxicity against primary tumor-associated macrophages within GBM tumors. The concept of targeting both tumor and immunosuppressive cells with MC9999 was further validated using CAR T cells derived from cancer patients. These findings establish MC9999 as a foundation for the development of effective CAR T cell therapies against solid tumors.
Collapse
Affiliation(s)
- Yan Luo
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Martha E. Gadd
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | - Yaqing Qie
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Mieu M. Brooks
- The Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
| | | | - Tanya Hundal
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | - Shuhua Li
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | | | - Yanyan Lou
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Tushar Patel
- Hepatology & Liver Transplantation, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Roxana Dronca
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamed A. Kharfan-Dabaja
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Alfredo Quinones-Hinojosa
- The Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Hong Qin
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
30
|
Lyukmanova EN, Kirichenko AV, Medyanik IA, Yashin KS, Kirpichnikov MP, Bychkov ML. Extracellular Vesicles from Plasma of Patients with Glioblastoma Promote Invasion of Glioblastoma Cells Even After Tumor Resection. Biomedicines 2024; 12:2834. [PMID: 39767739 PMCID: PMC11673896 DOI: 10.3390/biomedicines12122834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Glioblastoma (GB) is a highly aggressive tumor, whose progression is mediated by secretion of extracellular vesicles (EVs), which can pass the brain-blood barrier and be found in the plasma. Here, we performed a comparative analysis of the effects of EVs from the plasma of healthy donors (hEVs) and GB patients before (bEVs) and after (aEVs) tumor surgical resection on invasion of normal astrocytes and GB cells. Methods: We performed the transwell invasion assay, analyzed MAP kinases activation by Western blotting, studied SNAI1/SNAI2 cellular localization by confocal microscopy, measured cadherins expression by flow cytometry, and analyzed secretion of cytokines, which regulate migration and inflammation, by immunoassay. Results: hEVs did not affect invasion of astrocytes and GB cells, there was down-regulated cadherins expression in astrocytes, while there was increased E- and N-cadherin expression in GB cells. hEVs increased the secretion of inflammation and adhesion regulators both in astrocytes and GB cells. bEVs enhanced the invasion of GB cells but not of astrocytes via MAP AKT, JNK1/2/3, and p38 kinases activation, stimulated the clasterization of SNAI1 in the GB cell nucleus, promoted an E/N cadherin switch, and caused the secretion of inflammation and adhesion regulators in astrocytes and GB cells. aEVs exhibited the most of pro-oncogenic effects of bEVs (stimulation of GB cell invasion, SNAI1 nuclear localization, JNK1/2/3 activation, E/N cadherin switch, and secretion of inflammation and adhesion regulators in astrocytes and GB cells). However, aEVs effects were less pronounced than those of bEVs. Conclusions: In our study, we revealed common and different effects of plasma-derived hEVs, aEVs, and bEVs. hEVs can stimulate some pro-oncogenic effects in GB cells. Being less tumorigenic then bEVs, aEVs are still able to promote invasion of GB cells, probably remaining after tumor resection.
Collapse
Affiliation(s)
- Ekaterina N. Lyukmanova
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (A.V.K.); (M.P.K.)
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Artem V. Kirichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (A.V.K.); (M.P.K.)
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Igor A. Medyanik
- Department of Neurosurgery, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (I.A.M.); (K.S.Y.)
| | - Konstantin S. Yashin
- Department of Neurosurgery, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (I.A.M.); (K.S.Y.)
| | - Mikhail P. Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (A.V.K.); (M.P.K.)
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Maxim L. Bychkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia; (A.V.K.); (M.P.K.)
| |
Collapse
|
31
|
Yuan B, Gong H. General commentary: GQIcombi application to subdue glioma via differentiation therapy. Front Oncol 2024; 14:1466102. [PMID: 39726709 PMCID: PMC11669578 DOI: 10.3389/fonc.2024.1466102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- BinBin Yuan
- Department of Public Relations, Affiliated Haimen Hospital of Xinglin College, Nantong University, Nantong, Jiangsu, China
| | - Hui Gong
- Department of Neurosurgery, Affiliated Haimen Hospital of Xinglin College, Nantong
University, Nantong, Jiangsu, China
| |
Collapse
|
32
|
Zhou Y, Yao L, Ma T, Wang Z, Yin Y, Yang J, Zhang X, Zhang M, Qin G, Ma J, Zhao L, Liang J, Zhang J. Indoleamine 2,3-dioxygenase-1 involves in CD8 +T cell exhaustion in glioblastoma via regulating tryptophan levels. Int Immunopharmacol 2024; 142:113062. [PMID: 39244898 DOI: 10.1016/j.intimp.2024.113062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/01/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
Indoleamine 2,3-dioxygenase-1 (IDO-1) is an enzyme that catalyzes the metabolism of tryptophan (Trp). It is expressed in limited amounts in normal tissues but significantly upregulated during inflammation and infection. Various inflammatory factors, especially IFN-γ, can induce the expression of IDO-1. While extensive research has been conducted on the role of IDO-1 in tumors, its specific role in complex central nervous system tumors such as glioblastoma (GBM) remains unclear. This study aims to explore the role of IDO-1 in the development of GBM and analyze its association with tryptophan levels and CD8+T cell exhaustion in the tumor region. To achieve this, we constructed an orthotopic mouse glioblastoma tumor model to investigate the specific mechanisms between IDO-1, GBM, and CD8+T cell exhaustion. Our results showed that IDO-1 can promote CD8+T cell exhaustion by reducing tryptophan levels. When IDO-1 was knocked down in glioblastoma cells, other cells within the tumor microenvironment upregulated IDO-1 expression to compensate for the loss and enhance immunosuppressive effects. Therefore, the data suggest that the GBM microenvironment controls tryptophan levels by regulating IDO-1 expression, which plays a critical role in immune suppression. These findings support the use of immune therapy in combination with IDO-1 inhibitors or tryptophan supplementation as a potential treatment strategy.
Collapse
Affiliation(s)
- Yue Zhou
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Lina Yao
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Tingting Ma
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Zhongming Wang
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Yihe Yin
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Jian Yang
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Xuying Zhang
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Mingqi Zhang
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Gaofeng Qin
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Jinghan Ma
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Liang Zhao
- Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, Liaoning, China
| | - Jia Liang
- Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Liaoning Provincial Key Laboratory of Neurodegenerative Diseases and Department of Neurobiology, Jinzhou Medical University, China.
| | - Jinyi Zhang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranostics, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, Liaoning, China.
| |
Collapse
|
33
|
Duan J, Chen J, Lin Y, Lin SL, Wu J. Endocannabinoid Receptor 2 Function is Associated with Tumor-Associated Macrophage Accumulation and Increases in T Cell Number to Initiate a Potent Antitumor Response in a Syngeneic Murine Model of Glioblastoma. Cannabis Cannabinoid Res 2024; 9:1524-1536. [PMID: 38888628 PMCID: PMC11685299 DOI: 10.1089/can.2024.0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024] Open
Abstract
Introduction: Glioblastoma patients have a highly immunosuppressive tumor microenvironment and systemic immunosuppression that comprise a major barrier to immune checkpoint therapy. Based on the production of endocannabinoids by glioblastomas, we explored involvement of endocannabinoid receptor 2 (CB2R), encoded by the CNR2 gene, which is predominantly expressed by immune cells, in glioblastoma-related immunosuppression. Materials & Methods: Bioinformatics of human glioblastoma databases was used to correlate enzymes involved in the synthesis and degradation of endocannabinoids, as well as CB2Rs, with patient overall survival. Intrastriatal administration of luciferase-expressing, murine GL261 glioblastoma cells was used to establish in in vivo glioblastoma model for characterization of tumor growth and intratumoral immune cell infiltration, as well as provide immune cells for in vitro co-culture experiments. Involvement of CB2Rs was determined by treatment with CB2R agonist (GW405833) or CB2R antagonist (AM630). ELISA, FACS, and immunocytochemistry were used to determine perforin, granzyme B, and surface marker levels. Results: Bioinformatics of human glioblastoma databases showed high expression of CB2R and elevated endocannabinoid production correlated with poorer prognosis, and involved immune-associated pathways. AM630treatment of GL261 glioblastoma-bearing mice induced a potent antitumor response, with survival plateauing at 50% on Day 40, when all control mice (median survival 28 days) and mice treated with GW405833 (median survival 21 days) had died. Luciferase tumor imaging revealed accelerated tumor growth by GW405833 treatment, but stable or regressing tumors in AM630-treated mice. Notably, in spleens, AM630 treatment caused an 83% decrease in monocytes/macrophages, and 1.8- and 1.6-fold increases in CD8+ and CD4+ cells, respectively. Within tumors, there was a corresponding decrease in tumor-associated macrophages (TAMs) and increase in CD8+ T cells. In vitro, lymphocytes from AM630-treated mice showed greater cytotoxic function (increased percentage of perforin- and granzyme B-positive CD8+ T cells). Discussion: These results suggest that inhibition of CB2R enhances both immunosuppressive TAM infiltration and systemic T-cell suppression through CB2R activation, and that inhibition of CB2Rs can potently counter both the immunosuppressive tumor microenvironment, as well as systemic immunosuppression in glioblastoma.
Collapse
Affiliation(s)
- Jin Duan
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| | - Jieling Chen
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| | - Yilin Lin
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, Shantou, China
| | - Stanley L. Lin
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, Shantou, China
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, China
| | - Jie Wu
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| |
Collapse
|
34
|
YANG YUSHI, HU CHUJIAO, LEI SHAN, BAO XIN, ZENG ZHIRUI, CAO WENPENG. Using Multi-Omics Analysis to Explore Diagnostic Tool and Optimize Drug Therapy Selection for Patients with Glioma Based on Cross-Talk Gene Signature. Oncol Res 2024; 32:1921-1934. [PMID: 39574472 PMCID: PMC11576925 DOI: 10.32604/or.2024.046191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/28/2024] [Indexed: 11/24/2024] Open
Abstract
Background The heterogeneity of prognosis and treatment benefits among patients with gliomas is due to tumor microenvironment characteristics. However, biomarkers that reflect microenvironmental characteristics and predict the prognosis of gliomas are limited. Therefore, we aimed to develop a model that can effectively predict prognosis, differentiate microenvironment signatures, and optimize drug selection for patients with glioma. Materials and Methods The CIBERSORT algorithm, bulk sequencing analysis, and single-cell RNA (scRNA) analysis were employed to identify significant cross-talk genes between M2 macrophages and cancer cells in glioma tissues. A predictive model was constructed based on cross-talk gene expression, and its effect on prognosis, recurrence prediction, and microenvironment characteristics was validated in multiple cohorts. The effect of the predictive model on drug selection was evaluated using the OncoPredict algorithm and relevant cellular biology experiments. Results A high abundance of M2 macrophages in glioma tissues indicates poor prognosis, and cross-talk between macrophages and cancer cells plays a crucial role in shaping the tumor microenvironment. Eight genes involved in the cross-talk between macrophages and cancer cells were identified. Among them, periostin (POSTN), chitinase 3 like 1 (CHI3L1), serum amyloid A1 (SAA1), and matrix metallopeptidase 9 (MMP9) were selected to construct a predictive model. The developed model demonstrated significant efficacy in distinguishing patient prognosis, recurrent cases, and characteristics of high inflammation, hypoxia, and immunosuppression. Furthermore, this model can serve as a valuable tool for guiding the use of trametinib. Conclusions In summary, this study provides a comprehensive understanding of the interplay between M2 macrophages and cancer cells in glioma; utilizes a cross-talk gene signature to develop a predictive model that can predict the differentiation of patient prognosis, recurrence instances, and microenvironment characteristics; and aids in optimizing the application of trametinib in glioma patients.
Collapse
Affiliation(s)
- YUSHI YANG
- Department of Pathology and Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - CHUJIAO HU
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - SHAN LEI
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - XIN BAO
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - ZHIRUI ZENG
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - WENPENG CAO
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, 550025, China
| |
Collapse
|
35
|
Kuzminska J, Szyk P, Mlynarczyk DT, Bakun P, Muszalska-Kolos I, Dettlaff K, Sobczak A, Goslinski T, Jelinska A. Curcumin Derivatives in Medicinal Chemistry: Potential Applications in Cancer Treatment. Molecules 2024; 29:5321. [PMID: 39598712 PMCID: PMC11596437 DOI: 10.3390/molecules29225321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/20/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Curcumin, a naturally occurring compound found in the rhizome of Curcuma plants, particularly in turmeric (Curcuma longa L.), exhibits a broad range of biological activities, including anti-inflammatory, antioxidant, and anticancer properties. Curcumin has demonstrated effectiveness in inhibiting tumor growth, arousing interest for its potential in treating various cancers, such as breast, lung, prostate, and brain cancers. However, the clinical application of curcumin is limited due to its low chemical stability, poor water solubility, and low bioavailability. In response to these challenges, structural modifications of curcumin have been explored to improve its pharmacological properties, including enhanced anticancer selectivity index and bioavailability. This review highlights promising chemical modifications of curcumin that could lead to the development of more effective anticancer therapies. By functionalizing the parent curcumin molecule, researchers aim to create more stable and bioavailable compounds with enhanced therapeutic potential, making curcumin derivatives promising candidates for medical applications.
Collapse
Affiliation(s)
- Joanna Kuzminska
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland;
| | - Piotr Szyk
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland;
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Dariusz T. Mlynarczyk
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Pawel Bakun
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Izabela Muszalska-Kolos
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| | - Katarzyna Dettlaff
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| | - Agnieszka Sobczak
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| | - Tomasz Goslinski
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (D.T.M.); (P.B.)
| | - Anna Jelinska
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (I.M.-K.); (K.D.); (A.S.); (A.J.)
| |
Collapse
|
36
|
Wang L, Liang J, Ji S, Wang C, Huang Q. Potential Mechanism and Involvement of p120-Catenin in the Malignant Biology of Glioma. J Korean Neurosurg Soc 2024; 67:609-621. [PMID: 38956806 PMCID: PMC11540527 DOI: 10.3340/jkns.2024.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/19/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE This study analyzed the influence of p120-catenin (catenin [cadherin-associated protein], delta 1 [CTNND1]) on the malignant characteristics of glioma and elucidated the potential underlying mechanism. METHODS The p120 expression level was assessed in the brain tissues of 42 glioma patients and 10 patients with epilepsy by using the immunohistochemical method. Meanwhile, quantitative polymerase chain reaction (QT-PCR) technology was employed to assess the expression of p120 in the brain tissues of 71 glioma patients and 13 epilepsy patients. LN229, U251, and U87 glioma cells were used for in vitro analysis and categorized into four treatment groups : siRNA-blank control (BC) group (no RNA sequence was transfected), siRNA-negative control (NC) group (transfected control RNA sequences with no effect), and siRNA-1 and siRNA-2 groups (two p120-specific interfering RNA transfection). p120 expression in these treatment groups was quantified by western blotting assay. The migratory and invasive capabilities of glioma cells were studied by wound healing assay and Transwell invasion assay, respectively, under different treatment conditions. MTT (3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide) assay and cell cycle and apoptosis assay were used to determine glioma cell proliferation and apoptosis, respectively. Enzymelabeled assay was performed to measure intracellular calcium ion concentration. Immunofluorescence assay was performed for determining microtubule formation and glioma cell distribution. RESULTS Brain tissues of the glioma group exhibited a remarkable increase in the p120 expression level as compared to brain tissues of the nontumor group (p<0.05). Furthermore, a strong positive correlation was noted between the malignancy degree in glioma brain tissues and p120 expression in Western blotting (r=0.906, p<0.0001) and QT-PCR (F=830.6, p<0.01). Compared to the BC and NC groups, the siRNA transfection groups showed a significant suppression in p120 expression in glioma cells (p<0.05), with a marked attenuation in the invasive, migratory, and proliferative capabilities of glioma cells as well as an increase in apoptotic potential (p<0.05). Enzyme-labeled assay showed a remarkable increase in calcium concentration in glioma cells after siRNA treatment. Immunofluorescence assay revealed that the microtubule formation ability of glioma cells reduced after siRNA treatment. CONCLUSION p120 has a pivotal involvement in facilitating glioma cell invasion and proliferation by potentially modulating these processes through its involvement in microtubule formation and regulation of intracellular calcium ion levels.
Collapse
Affiliation(s)
- Leilei Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, China
| | - Jianshen Liang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Suzhen Ji
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Chunlou Wang
- Department of Pathology, Cangzhou Central Hospital, Cangzhou, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
37
|
Wei R, Xie H, Zhou Y, Chen X, Zhang L, Bui B, Liu X. VCAN in the extracellular matrix drives glioma recurrence by enhancing cell proliferation and migration. Front Neurosci 2024; 18:1501906. [PMID: 39554845 PMCID: PMC11565936 DOI: 10.3389/fnins.2024.1501906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Gliomas are the most prevalent primary malignant intracranial tumors, characterized by high rates of therapy resistance, recurrence, and mortality. A major factor contributing to the poor prognosis of gliomas is their ability to diffusely infiltrate surrounding and even distant brain tissues, rendering complete total resection almost impossible and leading to frequent recurrences. The extracellular matrix (ECM) plays a key role in the tumor microenvironment and may significantly influence glioma progression, recurrence, and therapeutic response. Methods In this study, we first identified the ECM and the Versican (VCAN), a key ECM protein, as critical contributors to glioma recurrence through a comprehensive analysis of transcriptomic data comparing recurrent and primary gliomas. Using single-cell sequencing, we revealed heterogeneous distribution patterns and extensive intercellular communication among ECM components. External sequencing and immunohistochemical (IHC) staining further validated that VCAN is significantly upregulated in recurrent gliomas and is associated with poor patient outcomes. Results Functional assays conducted in glioma cell lines overexpressing VCAN demonstrated that VCAN promotes cell proliferation and migration via the PI3K/Akt/AP-1 signaling pathway. Furthermore, inhibiting the PI3K/Akt pathway effectively blocked VCAN-mediated glioma progression. Conclusion These findings provide valuable insights into the mechanisms underlying glioma recurrence and suggest that targeting both VCAN and the PI3K/Akt pathway could represent a promising therapeutic strategy for managing recurrent gliomas.
Collapse
Affiliation(s)
- Ruolun Wei
- Department of Neurosurgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Haoyun Xie
- Biochemistry and Molecular Biology, Georgetown University, Washington, DC, United States
| | - Yukun Zhou
- Department of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xuhao Chen
- Department of Pathophysiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Liwei Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Brandon Bui
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
- Department of Human Biology, Stanford University, Stanford, CA, United States
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
38
|
Zhang L, Wang Y, Cai X, Mao X, Sun H. Deciphering the CNS-glioma dialogue: Advanced insights into CNS-glioma communication pathways and their therapeutic potential. J Cent Nerv Syst Dis 2024; 16:11795735241292188. [PMID: 39493257 PMCID: PMC11528668 DOI: 10.1177/11795735241292188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024] Open
Abstract
The field of cancer neuroscience has rapidly evolved, shedding light on the complex interplay between the nervous system and cancer, with a particular focus on the relationship between the central nervous system (CNS) and gliomas. Recent advancements have underscored the critical influence of CNS activity on glioma progression, emphasizing the roles of neurons and neuroglial cells in both the onset and evolution of gliomas. This review meticulously explores the primary communication pathways between the CNS and gliomas, encompassing neuro-glioma synapses, paracrine mechanisms, extracellular vesicles, tunneling nanotubes, and the integrative CNS-immune-glioma axis. It also evaluates current and emerging therapeutic interventions aimed at these pathways and proposes forward-looking perspectives for research in this domain.
Collapse
Affiliation(s)
- Lu Zhang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yajing Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxi Cai
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyuan Mao
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong–Hong Kong–Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Oldak L, Basa A, Milewska P, Chludzinska-Kasperuk S, Starosz A, Grubczak K, Kobus K, Reszec-Gielazyn J, Gorodkiewicz E. Preliminary studies on changes in the amount of tryptophan metabolites in human glioma tissues. Anal Chim Acta 2024; 1327:343149. [PMID: 39266061 DOI: 10.1016/j.aca.2024.343149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND We have developed and validated methods for the determination of three major tryptophan metabolites metabolized by the kynurenine pathway, namely kynurenine (KYN), 3-hydroxykynurenine (3-HK), and 3-hydroxyanthranilic acid (3-HAA). KYN and 3-HK were determined using RP-HPLC-UV, and 3-HAA using RP-HPLC-FL. We then developed a comparative method based on CE-UV. The developed methods were validated and 36 samples of human brain glioma tissue homogenates were assayed in all 4 grades of malignancy, and the concentration levels of assayed metabolites were compared with available clinical data. RESULTS Each of the methods is characterized by high precision, accuracy and repeatability, and the determined LOQ values indicate the possibility of performing quantitative analysis on the available samples of human glioma tumors (36 samples in grades G1-G4). The concentration values of selected metabolites obtained using HPLC methods were subjected to statistical analysis and preliminary clinical data processing. We found statistically significant differences in the concentrations of KYN, 3-HK and 3-HAA between the various grades of the disease, and characterized these differences more precisely by means of the Dunn-Bonferroni post hoc test. We did not find that the patient's environment or habits significantly affected the metabolites concentration of the study samples population. In addition, we showed a high positive correlation between KYN, 3-HK and 3-HAA, which appears to be a characteristic that describes metabolic changes of Trp in relation to KYN, 3-HK and 3-HAA, and indicates potential diagnostic value. SIGNIFICANCE The preliminary studies carried out contribute new knowledge on the molecular basis of human brain glioma. They also provide valuable information useful for the development of glioma diagnostics, differentiation of disease grades and assessment of the patient's condition. The obtained relationships between metabolite concentrations and the grade of malignancy of the disease and correlations between metabolite concentrations constitute the basis for further broader biochemical and clinical analysis.
Collapse
Affiliation(s)
- Lukasz Oldak
- Bioanalysis Laboratory, Department of Physical Chemistry, Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245, Bialystok, Poland.
| | - Anna Basa
- Department of Materials Chemistry, Department of Physical Chemistry, Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245, Bialystok, Poland.
| | - Patrycja Milewska
- Biobank, Biobank at Medical University of Bialystok, Waszyngtona 13, 15-269, Bialystok, Poland.
| | | | - Aleksandra Starosz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Waszyngtona 13, 15-269, Bialystok, Poland.
| | - Kamil Grubczak
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Waszyngtona 13, 15-269, Bialystok, Poland.
| | - Krzysztof Kobus
- Private Surgical Clinic in Bialystok, Dr. Kobus Chirurgia, Fabryczna 20, 15-482, Poland.
| | - Joanna Reszec-Gielazyn
- Biobank, Biobank at Medical University of Bialystok, Waszyngtona 13, 15-269, Bialystok, Poland; Department of Medical Pathology, Medical University of Bialystok, Waszyngtona 13, 15-269, Bialystok, Poland.
| | - Ewa Gorodkiewicz
- Bioanalysis Laboratory, Department of Physical Chemistry, Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245, Bialystok, Poland.
| |
Collapse
|
40
|
Chen C, Yuan M, Xia L, Wu X, Zhong X, Zhang H, Zhang L, Liu X, Wang Z, Sun C. Expression of CREBBP and EP300 Associated With Tumor Volume in Patients With Grade-3 Glioma: A Retrospective Analysis. Clin Med Insights Oncol 2024; 18:11795549241287777. [PMID: 39429683 PMCID: PMC11490948 DOI: 10.1177/11795549241287777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024] Open
Abstract
Background Reliable predictive data are crucial for making accurate treatment decisions in glioma patients, but it can be challenging to obtain due to limited information in many cases. Numerous research studies have indicated the involvement of cyclic adenosine monophosphate (cAMP)-response element binding protein (CREBBP) and E1A binding protein p300 (EP300) in tumorigenesis and tumor progression across various types. Methods The messenger RNA (mRNA) expression levels of CREBBP and EP300 were retrospectively analyzed in 17 grade-3 glioma patients. The SYBR Green real-time polymerase chain reaction (RT-PCR) technique was employed for mRNA expression analysis, with the glyceraldehyde-3-phosphate dehydrogenase gene (GAPDH) used as a reference gene for data normalization. In addition, the relationship between CREBBP, EP300 expression and patients' clinical information, imaging features, histologic features, immune factors, and overall survival was assessed through univariate analyses. Results The analysis of the data unveiled a statistically significant upregulation of CREBBP and EP300 mRNA expression levels in large gliomas as compared with their smaller counterparts (P < .05). Histological examination using hematoxylin and eosin (H&E) staining exhibited marked cellular heterogeneity, with heightened cell density observed specifically within tumors displaying elevated CREBBP expression levels. In contrast, there was a substantial downregulation of complement 3 and complement 4 within larger tumor volumes when compared with smaller ones (P < .05). However, these findings do not serve as clinically relevant prognostic indicators for glioma. Conclusions It is suggested that higher expression levels of CREBBP and EP300 are positively associated with increased tumor volume. Inhibition of CREBBP and EP300 enhances local immunogenicity, leading to the recruitment of immune cells and release of cytokines for effective tumor eradication, ultimately resulting in the inhibition of tumor growth.
Collapse
Affiliation(s)
- Cuiwei Chen
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Meiqin Yuan
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Xia
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xin Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingguang Zhong
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Huangjie Zhang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Lidan Zhang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xuan Liu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Zeng Wang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Integrated Traditional Chinese and Western Medicine Oncology Laboratory, Key Laboratory of Traditional Chinese Medicine of Zhejiang Province, Hangzhou, China
| | - Caixing Sun
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
41
|
Li D, Zhang Z, Wang L. Emerging role of tumor microenvironmental nutrients and metabolic molecules in ferroptosis: Mechanisms and clinical implications. Biomed Pharmacother 2024; 179:117406. [PMID: 39255738 DOI: 10.1016/j.biopha.2024.117406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
In recent years, ferroptosis has gradually attracted increasing attention because of its important role in tumors. Ferroptosis resistance is an important cause of tumor metastasis, recurrence and drug resistance. Exploring the initiating factors and specific mechanisms of ferroptosis has become a key strategy to block tumor progression and improve drug sensitivity. As the external space in direct contact with tumor cells, the tumor microenvironment has a great impact on the biological function of tumor cells. The relationships between abnormal environmental characteristics (hypoxia, lactic acid accumulation, etc.) in the microenvironment and ferroptosis of tumor cells has not been fully characterized. This review focuses on the characteristics of the tumor microenvironment and summarizes the mechanisms of ferroptosis under different environmental factors, aiming to provide new insights for subsequent targeted therapy. Moreover, considering the presence of anticancer drugs in the microenvironment, we further summarize the mechanisms of ferroptosis to provide new strategies for the sensitization of tumor cells to drugs.
Collapse
Affiliation(s)
- Dongyu Li
- Department of VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Wang
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
42
|
Tataranu LG, Turliuc S, Kamel A, Rizea RE, Dricu A, Staicu GA, Baloi SC, Rodriguez SMB, Manole AIM. Glioblastoma Tumor Microenvironment: An Important Modulator for Tumoral Progression and Therapy Resistance. Curr Issues Mol Biol 2024; 46:9881-9894. [PMID: 39329940 PMCID: PMC11430601 DOI: 10.3390/cimb46090588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The race to find an effective treatment for glioblastoma (GBM) remains a critical topic, because of its high aggressivity and impact on survival and the quality of life. Currently, due to GBM's high heterogeneity, the conventional treatment success rate and response to therapy are relatively low, with a median survival rate of less than 20 months. A new point of view can be provided by the comprehension of the tumor microenvironment (TME) in pursuance of the development of new therapeutic strategies to aim for a longer survival rate with an improved quality of life and longer disease-free interval (DFI). The main components of the GBM TME are represented by the extracellular matrix (ECM), glioma cells and glioma stem cells (GSCs), immune cells (microglia, macrophages, neutrophils, lymphocytes), neuronal cells, all of them having dynamic interactions and being able to influence the tumoral growth, progression, and drug resistance thus being a potential therapeutic target. This paper will review the latest research on the GBM TME and the potential therapeutic targets to form an up-to-date strategy.
Collapse
Affiliation(s)
- Ligia Gabriela Tataranu
- Neurosurgical Department, University of Medicine and Pharmacy "Carol Davila", 020022 Bucharest, Romania
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy "G. T. Popa", 700115 Iasi, Romania
| | - Amira Kamel
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Radu Eugen Rizea
- Neurosurgical Department, University of Medicine and Pharmacy "Carol Davila", 020022 Bucharest, Romania
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | | | - Stefania Carina Baloi
- Biochemistry Department, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | | | | |
Collapse
|
43
|
An W, Ren C, Yuan L, Qiu Z, Wang P, Cheng Y, He Z, Han X, Li S, An Y. High expression of SIGLEC7 may promote M2-type macrophage polarization leading to adverse prognosis in glioma patients. Front Immunol 2024; 15:1411072. [PMID: 39211050 PMCID: PMC11357930 DOI: 10.3389/fimmu.2024.1411072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Gliomas are the most common primary intracranial tumors, known for their high invasiveness and destructiveness. Sialic acid-binding immunoglobulin-like lectin 7 (SIGLEC7) is present in various immune cells, especially macrophages, and significantly affects immune homeostasis and cancer cell response. However, research on the role and prognostic impact of SIGLEC7 in glioma patients is currently limited. Methods We utilized transcriptomic data from 702 glioma patients in The Cancer Genome Atlas (TCGA) and 693 glioma patients in the Chinese Glioma Genome Atlas (CGGA), along with clinical samples we collected, to comprehensively investigate the impact of SIGLEC7 on glioma expression patterns, biological functions, and prognostic value. We focused on its role in glioma-related immune responses and immune cell infiltration and analyzed its expression at the single-cell level. Finally, we validated the role of SIGLEC7 in gliomas through tissue and cell experiments. Results SIGLEC7 expression was significantly increased in glioma patients with malignant characteristics. Survival analysis indicated that glioma patients with high SIGLEC7 expression had significantly lower survival rates. Gene function analysis revealed that SIGLEC7 is primarily involved in immune and inflammatory responses and is strongly negatively correlated with tumor-associated immune regulation. Additionally, the expression of most immune checkpoints was positively correlated with SIGLEC7, and immune cell infiltration analysis clearly demonstrated a significant positive correlation between SIGLEC7 expression and M2 macrophage infiltration levels. Single-cell analysis, along with tissue and cell experiments, confirmed that SIGLEC7 enhances macrophage polarization towards the M2 phenotype, thereby promoting glioma invasiveness through the immunosuppressive effects of M2 macrophages. Cox regression analysis and the establishment of survival prediction models indicated that high SIGLEC7 expression is an unfavorable prognostic factor for glioma patients. Discussion High SIGLEC7 expression predicts poor prognosis in glioma patients and is closely associated with M2 macrophages in the tumor environment. In the future, SIGLEC7 may become a promising target for glioma immunotherapy.
Collapse
Affiliation(s)
- Wenhao An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Changyuan Ren
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Lei Yuan
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Zhiqiang Qiu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, United States
| | - Peishen Wang
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Yanwen Cheng
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Zi He
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Xinye Han
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yihua An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Zhang X, Zhang X, Liu T, Sha K. Comprehensive analysis of the prognostic and immunological signature of TNFAIP8 family genes in human glioma. Sci Rep 2024; 14:17875. [PMID: 39090168 PMCID: PMC11294591 DOI: 10.1038/s41598-024-68784-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
TNFAIP8 family molecules have been recognized for their involvement in the progression of tumors across a range of cancer types. Emerging experimental data suggests a role for certain TNFAIP8 family molecules in the development of glioma. Nonetheless, the comprehensive understanding of the genomic alterations, prognostic significance, and immunological profiles of TNFAIP8 family molecules in glioma remains incomplete. In the study, using the comprehensive bioinformatics tools, we explored the unique functions of 4 TNFAIP8 members including TNFAIP8, TNFAIP8L1, TNFAIP8L2 and TNFAIP8L3 in glioma. The expressions of TNFAIP8, TNFAIP8L1, TNFAIP8L2, and TNFAIP8L3 were notably upregulated in glioma tissues compared to normal tissues. Furthermore, survival analysis indicated that elevated expression levels of TNFAIP8, TNFAIP8L1 and TNFAIP8L2 were correlated with unfavorable outcomes in terms of overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) among glioma patients. Genetic modifications, such as mutations and copy number alterations, within the TNFAIP8 family exhibited a significant association with extended OS, DSS and PFS in individuals diagnosed with glioma. The findings suggest a noteworthy correlation between TNFAIP8 family members and the age and 1p/19q codeletion status of glioma patients. We also found that there were significant relationships between TNFAIP8 family expression and tumor immunity in glioma. Furthermore, functional annotation of TNFAIP8 family members and their co-expressed genes in gliomas was carried out using GO and KEGG pathway analysis. The GO analysis revealed that the primary biological processes influenced by the TNFAIP8 family co-expressed genes included cell chemotaxis, temperature homeostasis, and endocytic vesicle formation. Additionally, the KEGG analysis demonstrated that TNFAIP8 family co-expressed genes are involved in regulating various pathways such as inflammatory mediator regulation of TRP channels, pathways in cancer, prolactin signaling pathway, and Fc gamma R-mediated phagocytosis. Overall, the findings suggest that TNFAIP8 family members may play a significant role in the development of glioma and have the potential to serve as prognostic indicators and therapeutic targets for individuals with glioma.
Collapse
Affiliation(s)
- Xuezhong Zhang
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, China
| | - Xuebin Zhang
- Department of Anorectal Surgery, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, China
| | - Tonggang Liu
- Department of Infectious Diseases, Binzhou Medical University Hospital, Binzhou, 256603, Shandong, China.
| | - Kaihui Sha
- Binzhou Medical University School of Nursing, Binzhou, 256603, Shandong, China.
| |
Collapse
|
45
|
Yang X, Niu W, Wu K, Li X, Hou H, Tan Y, Wang X, Yang G, Wang L, Zhang H. Diffusion kurtosis imaging-based habitat analysis identifies high-risk molecular subtypes and heterogeneity matching in diffuse gliomas. Ann Clin Transl Neurol 2024; 11:2073-2087. [PMID: 38887966 PMCID: PMC11330218 DOI: 10.1002/acn3.52128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/14/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE High-risk types of diffuse gliomas in adults include isocitrate dehydrogenase (IDH) wild-type glioblastomas and grade 4 astrocytomas. Achieving noninvasive prediction of high-risk molecular subtypes of gliomas is important for personalized and precise diagnosis and treatment. METHODS We retrospectively collected data from 116 patients diagnosed with adult diffuse gliomas. Multiple high-risk molecular markers were tested, and various habitat models and whole-tumor models were constructed based on preoperative routine and diffusion kurtosis imaging (DKI) sequences to predict high-risk molecular subtypes of gliomas. Feature selection and model construction utilized Least absolute shrinkage and selection operator (LASSO) and support vector machine (SVM). Finally, the Wilcoxon rank-sum test was employed to explore the correlation between habitat quantitative features (intra-tumor heterogeneity score,ITH score) and heterogeneity, as well as high-risk molecular subtypes. RESULTS The results showed that the habitat analysis model based on DKI performed remarkably well (with AUC values reaching 0.977 and 0.902 in the training and test sets, respectively). The model's performance was further enhanced when combined with clinical variables. (The AUC values were 0.994 and 0.920, respectively.) Additionally, we found a close correlation between ITH score and heterogeneity, with statistically significant differences observed between high-risk and non-high-risk molecular subtypes. INTERPRETATION The habitat model based on DKI is an ideal means for preoperatively predicting high-risk molecular subtypes of gliomas, holding significant value for noninvasively alerting malignant gliomas and those with malignant transformation potential.
Collapse
Affiliation(s)
- Xiangli Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalTaiyuan030032China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Wenju Niu
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Kai Wu
- Department of Information ManagementFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiang Li
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Heng Hou
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Yan Tan
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiaochun Wang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Guoqiang Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Lei Wang
- Beijing Tiantan HospitalCapital Medical UniversityBeijing100050China
| | - Hui Zhang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Intelligent Imaging Big Data and Functional Nano‐imaging Engineering Research Center of Shanxi ProvinceFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
46
|
Li M, Sun P, Tu B, Deng G, Li D, He W. Hypoxia conduces the glioma progression by inducing M2 macrophage polarization via elevating TNFSF9 level in a histone-lactylation-dependent manner. Am J Physiol Cell Physiol 2024; 327:C487-C504. [PMID: 39010835 DOI: 10.1152/ajpcell.00124.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Hypoxia is a critical factor contributing to a poor prognosis and challenging glioma therapy. Previous studies have indicated that hypoxia drives M2 polarization of macrophages and promotes cancer progression in various solid tumors. However, the more complex and diverse mechanisms underlying this process remain to be elucidated. Here, we aimed to examine the functions of hypoxia in gliomas and preliminarily investigate the underlying mechanisms of M2 macrophage polarization caused by hypoxia. We found that hypoxia significantly enhances the malignant phenotypes of U87 and U251 cells by regulating glycolysis. In addition, hypoxia mediated accumulation of the glycolysis product [lactic acid (LA)], which is subsequently absorbed by macrophages to induce its M2 polarization, and this process is reverted by both the glycolysis inhibitor and silenced monocarboxylate transporter (MCT-1) in macrophages, indicating that M2 macrophage polarization is associated with the promotion of glycolysis by hypoxia. Interestingly, we also found that hypoxia mediated LA accumulation in glioma cells upon uptake by macrophages upregulates H3K18La expression and promotes tumor necrosis factor superfamily member 9 (TNFSF9) expression in a histone-lactylation-dependent manner based on the results of chromatin immunoprecipitation sequencing (ChIP seq) enrichment analysis. Subsequent in vitro and in vivo experiments further indicated that TNFSF9 facilitated glioma progression. Mechanistically, hypoxia-mediated LA accumulation in glioma cells is taken up by macrophages and then induces its M2 macrophage polarization by regulating TNFSF9 expression via MCT-1/H3K18La signaling, thus facilitating the malignant progression of gliomas.NEW & NOTEWORTHY Our study revealed that hypoxia induces the production of LA accumulation through glycolysis in glioma cells, which is subsequently absorbed by macrophages and leads to its M2 polarization via the MCT-1/H3K18La/TNFSF9 axis, ultimately significantly promoting the malignant progression of glioma cells. These findings are novel and noteworthy as they provide insights into the connection between energy metabolism and epigenetics in gliomas.
Collapse
Affiliation(s)
- Min Li
- Neurosurgery Department of Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, People's Republic of China
- Jiangxi Key Laboratory of Translational Research for Cancer, Nanchang, Jiangxi, People's Republic of China
| | - Pingfeng Sun
- Jiangxi Provincial Maternal and Child Health Care Hospital, Nanchang, Jiangxi, People's Republic of China
| | - Binfeng Tu
- Neurosurgery Department of Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, People's Republic of China
| | - Guojun Deng
- Neurosurgery Department of Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, People's Republic of China
| | - Donghai Li
- Neurosurgery Department of Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, People's Republic of China
| | - Wei He
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
47
|
Ma Y, Yi J, Ruan J, Ma J, Yang Q, Zhang K, Zhang M, Zeng G, Jin L, Huang X, Li J, Yang H, Wu W, Sun D. Engineered Cell Membrane-Coated Nanoparticles: New Strategies in Glioma Targeted Therapy and Immune Modulation. Adv Healthc Mater 2024; 13:e2400514. [PMID: 38652681 DOI: 10.1002/adhm.202400514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Gliomas, the most prevalent primary brain tumors, pose considerable challenges due to their heterogeneity, intricate tumor microenvironment (TME), and blood-brain barrier (BBB), which restrict the effectiveness of traditional treatments like surgery and chemotherapy. This review provides an overview of engineered cell membrane technologies in glioma therapy, with a specific emphasis on targeted drug delivery and modulation of the immune microenvironment. This study investigates the progress in engineered cell membranes, encompassing physical, chemical, and genetic alterations, to improve drug delivery across the BBB and effectively target gliomas. The examination focuses on the interaction of engineered cell membrane-coated nanoparticles (ECM-NPs) with the TME in gliomas, emphasizing their potential to modulate glioma cell behavior and TME to enhance therapeutic efficacy. The review further explores the involvement of ECM-NPs in immunomodulation techniques, highlighting their impact on immune reactions. While facing obstacles related to membrane stability and manufacturing scalability, the review outlines forthcoming research directions focused on enhancing membrane performance. This review underscores the promise of ECM-NPs in surpassing conventional therapeutic constraints, proposing novel approaches for efficacious glioma treatment.
Collapse
Affiliation(s)
- Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Maolan Zhang
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Guoming Zeng
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Xiaobei Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Haifeng Yang
- JinFeng Laboratory, Chongqing, 401329, China
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, 400044, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
48
|
Tataranu LG, Turliuc S, Rizea RE, Dricu A, Alexandru O, Staicu GA, Kamel A. A Synopsis of Biomarkers in Glioblastoma: Past and Present. Curr Issues Mol Biol 2024; 46:6903-6939. [PMID: 39057054 PMCID: PMC11275428 DOI: 10.3390/cimb46070412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Accounting for 48% of malignant brain tumors in adults, glioblastoma has been of great interest in the last decades, especially in the biomolecular and neurosurgical fields, due to its incurable nature and notable neurological morbidity. The major advancements in neurosurgical technologies have positively influenced the extent of safe tumoral resection, while the latest progress in the biomolecular field of GBM has uncovered new potential therapeutical targets. Although GBM currently has no curative therapy, recent progress has been made in the management of this disease, both from surgical and molecular perspectives. The main current therapeutic approach is multimodal and consists of neurosurgical intervention, radiotherapy, and chemotherapy, mostly with temozolomide. Although most patients will develop treatment resistance and tumor recurrence after surgical removal, biomolecular advancements regarding GBM have contributed to a better understanding of this pathology and its therapeutic management. Over the past few decades, specific biomarkers have been discovered that have helped predict prognosis and treatment responses and contributed to improvements in survival rates.
Collapse
Affiliation(s)
- Ligia Gabriela Tataranu
- Neurosurgical Department, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania;
- Neurosurgical Department, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy “G. T. Popa”, 700115 Iasi, Romania;
| | - Radu Eugen Rizea
- Neurosurgical Department, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania;
- Neurosurgical Department, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anica Dricu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy, 200349 Craiova, Romania (O.A.); (G.-A.S.)
| | - Oana Alexandru
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy, 200349 Craiova, Romania (O.A.); (G.-A.S.)
| | - Georgiana-Adeline Staicu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy, 200349 Craiova, Romania (O.A.); (G.-A.S.)
| | - Amira Kamel
- Neurosurgical Department, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| |
Collapse
|
49
|
Huang Q, Liang H, Shi S, Ke Y, Wang J. Identification of TNFAIP6 as a reliable prognostic indicator of low-grade glioma. Heliyon 2024; 10:e33030. [PMID: 38948040 PMCID: PMC11211890 DOI: 10.1016/j.heliyon.2024.e33030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/02/2024] Open
Abstract
Glioma is the most common primary malignant tumor in the brain, characterizing by high disability rate and high recurrence rate. Although low-grade glioma (LGG) has a relative benign biological behavior, the prognosis of LGG patients still varies greatly. Glioma stem cells (GSCs) are considered as the chief offenders of glioma cell proliferation, invasion and resistance to therapies. Our study screened a series of glioma stem cell-related genes (GSCRG) based on mDNAsi and WCGNA, and finally established a reliable single-gene prognostic model through 101 combinations of 10 machine learning methods. Our result suggested that the expression level of TNFAIP6 is negatively correlated with the prognosis of LGG patients, which may be the result of pro-cancer signaling pathways activation and immunosuppression. In general, this study revealed that TNFAIP6 is a robust and valuable prognostic factor in LGG, and may be a new target for LGG treatment.
Collapse
Affiliation(s)
| | | | - Shenbao Shi
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiquan Ke
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jihui Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
50
|
Kafka A, Pećina-Šlaus N, Drmić D, Bukovac A, Njirić N, Žarković K, Jakovčević A. SFRP4 protein expression is reduced in high grade astrocytomas which is not caused by the methylation of its promoter. Front Mol Neurosci 2024; 17:1398872. [PMID: 38993819 PMCID: PMC11236799 DOI: 10.3389/fnmol.2024.1398872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction Epigenetics play a vital role in stratifying CNS tumors and gliomas. The importance of studying Secreted frizzled-related protein 4 (SFRP4) in gliomas is to improve diffuse glioma methylation profiling. Here we examined the methylation status of SFRP4 promoter and the level of its protein expression in diffuse gliomas WHO grades 2-4. Methods SFRP4 expression was detected by immunohistochemistry and evaluated semi-quantitatively. In the tumor hot-spot area, the intensity of protein expression in 200 cells was determined using ImageJ (National Institutes of Health, United States). The assessment of immunopositivity was based on the IRS score (Immunoreactivity Score). Promoter methylation was examined by methylation specific-PCR (MSP) in fifty-one diffuse glioma samples and appropriate controls. Isolated DNA was treated with bisulfite conversion and afterwards used for MSP. Public databases (cBioPortal, COSMIC and LOVD) were searched to corroborate the results. Results and discussion SFRP4 protein expression in glioblastomas was very weak or non-existent in 86.7% of samples, moderate in 13.3%, while strong expression was not observed. The increase in astrocytoma grade resulted in SFRP4 protein decrease (p = 0.008), indicating the loss of its antagonistic role in Wnt signaling. Promoter methylation of SFRP4 gene was found in 16.3% of cases. Astrocytomas grade 2 had significantly more methylated cases compared to grade 3 astrocytomas (p = 0.004) and glioblastomas (p < 0.001), which may indicate temporal niche of methylation in grade 2. Furthermore, the expression levels of SFRP4 were high in samples with methylated SFRP4 promoter and low or missing in unmethylated cases (Pearson's R = -0.413; p = 0.003). We also investigated the association of SFRP4 changes to key Wnt regulators GSK3β and DKK3 and established a positive correlation between methylations of SFRP4 and GSK3β (Pearson's R = 0.323; p = 0.03). Furthermore, SFRP4 expression was correlated to unmethylated DKK3 (Chi square = 7.254; p = 0.027) indication that Wnt signaling antagonist is associated to negative regulator's demethylation. Conclusion The study contributes to the recognition of the significance of epigenetic changes in diffuse glioma indicating that restoring SFRP4 protein holds potential as therapeutic avenue. Reduced expression of SFRP4 in glioblastomas, not following promoter methylation pattern, suggests another mechanism, possible global methylation, that turns off SFRP4 expression in higher grades.
Collapse
Affiliation(s)
- Anja Kafka
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nives Pećina-Šlaus
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Denis Drmić
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anja Bukovac
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Niko Njirić
- Department of Neurosurgery, University Hospital Center “Zagreb”, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Kamelija Žarković
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Division of Pathology, University Hospital Center “Zagreb”, Zagreb, Croatia
| | - Antonia Jakovčević
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Division of Pathology, University Hospital Center “Zagreb”, Zagreb, Croatia
| |
Collapse
|