1
|
Liu H, Tan S, Li Z, Qi J, Tang X, Zhang J. OTUB1 promotes glioma progression by stabilizing TRAF4. Cell Signal 2025; 131:111704. [PMID: 40090557 DOI: 10.1016/j.cellsig.2025.111704] [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: 11/03/2024] [Revised: 02/17/2025] [Accepted: 02/25/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Glioma is a highly heterogeneous brain tumor with poor prognosis. This study aims to investigate the functional role of OTUB1 in glioma and its impact on TRAF4 stability, seeking potential therapeutic targets. METHODS We mined single-cell sequencing data from 12 glioma patients to analyze the heterogeneity of 20,145 glioma cells. The expression of OTUB1 in glioma tissues and cell lines was assessed using Western blot and qPCR. Additionally, immunoprecipitation and ubiquitination assays were conducted to evaluate the effect of OTUB1 on TRAF4 and its role in regulating TRAF4 stability. In vitro assays were performed to assess the effects of OTUB1 on cell proliferation, migration, and clonogenicity, while in vivo experiments using xenograft models in nude mice validated the impact of OTUB1 on tumor growth. RESULTS OTUB1 was found to be significantly overexpressed in glioma tissues, correlating with poor patient outcomes. Knockdown of OTUB1 markedly inhibited the proliferation and migration of LN229 and U87MG cells while increasing apoptosis. Immunoprecipitation studies revealed that OTUB1 stabilizes TRAF4 by inhibiting its ubiquitination, thereby promoting glioma cell proliferation and invasion. In vivo, tumors with OTUB1 knockdown demonstrated significantly reduced growth rates. CONCLUSION OTUB1 plays a critical role in glioma progression and may serve as a novel therapeutic target. The development of inhibitors targeting OTUB1 could potentially improve outcomes for glioma patients.
Collapse
Affiliation(s)
- Hongjun Liu
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shasha Tan
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zhou Li
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Jian Qi
- Department of Neurosurgery, Nanchong Fifth People's Hospital, Nanchong, Sichuan, China
| | - Xiaoping Tang
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China.
| | - Junhao Zhang
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
2
|
Wang Q, Zhang Y, Ma K, Lin P, Wang Y, Wang R, Li H, Li Z, Wang G. Plexin B2 in physiology and pathophysiology of the central nervous system. Int Immunopharmacol 2025; 155:114627. [PMID: 40220620 DOI: 10.1016/j.intimp.2025.114627] [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: 02/10/2025] [Revised: 04/05/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
The Plexin protein family was initially found in 1995, comprising subfamilies from Plexin A to Plexin D. Plexin B2, a member of the Plexin subfamily, has widespread expression in many human organs and tissues, particularly in the nervous system where expression levels are significantly heightened. The biological roles of Plexin B2 are mostly determined by its protein structure and functional domains. These domains regulate the binding selectivity and affinity for ligands. Ligand binding activates signal transduction pathways, resulting in regulatory effects on several biological processes. This includes managing brain growth and change, keeping angiogenesis and vascular homeostasis in check, and preventing the start, growth, and metastasis of cancer. Plexin B2 has also been associated with the onset of many nervous system illnesses. Plexin B2 aids in the invasion and spread of malignant cells, facilitates nerve healing following spinal cord damage, and plays a role in the etiology of schizophrenia. This article thoroughly examines the existing research on Plexin B2 and its importance in central nervous system biology. Simultaneously, it investigates the regulatory function of Plexin B2 across many cell types in the central nervous system, specifically neural stem cells, neurons, microglia, and astrocytes. This study examines the current knowledge of Plexin B2's role in central nervous system diseases, including schizophrenia, spinal cord injury, neuroblastoma, and fear memory. Overall, the prospects for the clinical translation of Plexin B2 are promising. However, challenges related to specificity and drug delivery must be addressed. Future research could explore the integration of nanodrug delivery systems to enhance the clinical application of Plexin B2-targeted therapies.
Collapse
Affiliation(s)
- Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yuan Zhang
- Department of Pharmacy, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai 200120, China
| | - Kaixuan Ma
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Peng Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Ran Wang
- School of Pharmacy, Harbin Medical University, Daqing, Heilongjiang 163319, China
| | - He Li
- Department of Parasitology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China.
| | - Guangtian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China; Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
3
|
Ljubimov VA, Sun T, Wang J, Li L, Wang PZ, Ljubimov AV, Holler E, Black KL, Kopeček J, Ljubimova JY, Yang J. Blood-brain barrier crossing biopolymer targeting c-Myc and anti-PD-1 activate primary brain lymphoma immunity: Artificial intelligence analysis. J Control Release 2025; 381:113611. [PMID: 40088978 DOI: 10.1016/j.jconrel.2025.113611] [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: 12/29/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Primary Central Nervous System Lymphoma is an aggressive central nervous system neoplasm with poor response to pharmacological treatment, partially due to insufficient drug delivery across blood-brain barrier. In this study, we developed a novel therapy for this lymphoma by combining a targeted nanopolymer treatment with an immune checkpoint inhibitor antibody (anti-PD-1). A N-(2-hydroxypropyl)methacrylamide copolymer-based nanoconjugate was designed to block tumor cell c-Myc oncogene expression by antisense oligonucleotide. Angiopep-2 peptide was conjugated to the copolymer to facilitate nanodrug crossing of the blood-brain barrier. Systemically administered polymeric nanodrug, alone or in combination with immune checkpoint inhibitor antibody anti-PD-1, was tested in syngeneic mouse model of A20 intracranial brain lymphoma. There was no significant survival difference between saline- and free anti-PD-1-treated groups. However, significant survival advantage vs. saline was observed upon treatment with nanodrug bearing Angiopep-2, H6 (6 histidines for endosome escape), and c-Myc antisense alone and especially when it was combined with anti-PD-1 antibody. Animal survival after combined treatment was also significantly increased vs. free anti-PD-1. Artificial Intelligence-assisted analysis of gene expression database after RNA-seq of tumors was used to find novel immune pathways, molecular targets and the most effective multifunctional drugs together with future drug prediction for brain lymphoma in vivo model. Spectral flow cytometry and RNA-seq analysis revealed a robust activation of tumor infiltrating T lymphocytes with enhanced interferon γ signaling and polarization to M1-type macrophages in treated tumors, which was confirmed by immunofluorescence staining. In summary, a new effective blood-brain barrier crossing nano immuno therapeutic system was developed that effectively blocked tumor c-Myc acting in combination with immune checkpoint inhibitor anti-PD-1 to treat primary brain lymphoma. The treatment improved survival of tumor-bearing animals through activation of both the adaptive and innate immune responses.
Collapse
Affiliation(s)
- Vladimir A Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States
| | - Jiawei Wang
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States
| | - Lian Li
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States
| | - Paul Z Wang
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Alexander V Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States; Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States; Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Eggehard Holler
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, United States; Institut für Biophysik und Physikalische Biochemie Universität Regensburg, D-93040 Regensburg, Germany
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States; Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Jindřich Kopeček
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States
| | - Julia Y Ljubimova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, United States.
| | - Jiyuan Yang
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States.
| |
Collapse
|
4
|
German-Cortés J, Herrero R, Torroglosa N, Pumarola A, Fischer-Albiol N, Campos-Moreno S, Sabaté S, Alcina À, Mancilla S, García B, Llaguno-Munive M, Díaz-Riascos ZV, Martins C, Schwartz S, Ferrer-Costa R, Abasolo I, Sánchez-Gómez P, Sarmento B, Rafael D, Andrade F. Preclinical evaluation of several polymeric micelles identifies Soluplus®-docetaxel as the most effective candidate in multiple glioblastoma models. J Control Release 2025; 381:113616. [PMID: 40073942 DOI: 10.1016/j.jconrel.2025.113616] [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: 11/10/2024] [Revised: 01/23/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal cancers, with limited treatment options due to the blood-brain barrier (BBB), systemic toxicity, and treatment resistance. Nanomedicine offers potential solutions to these challenges. This study explores Pluronic® F127 and Soluplus®-based micelles as carriers for Lomustine, Gefitinib, and Docetaxel to determine the optimal system for GBM therapy. Micelles were physicochemically characterized and biologically validated using U87-MG and U251-MG GBM cell lines in 2D and 3D models, assessing internalization, safety, and therapeutic efficacy. Soluplus® micelles (SM) showed favorable properties for intravenous administration, including low polydispersity, efficient drug release in the tumoral microenvironment, minimal cell toxicity, and a BBB-crossing rate of 15 %. Among the drugs tested, Docetaxel showed the lowest IC50 values in both 2D cell models and demonstrated superior efficacy in 3D cultures when delivered by SM. Molecular analysis confirmed that SM-D impacts key GBM-related pathways, affecting markers like E-cadherin, EPCAM, L1CAM, or EGFR. In vivo, SM-D significantly reduced tumor mass and cancer cell density, showing a favorable safety profile compared to free Docetaxel, as evidenced by reduced weight loss and histological assessments. Overall, SM-D stands out as the most promising approach for GBM treatment, supporting the potential of nanomedicine in overcoming the barriers to effective glioblastoma therapy.
Collapse
Affiliation(s)
- Júlia German-Cortés
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Doctoral program in Biotechnology, Faculty of Pharmacy and Food Sciences, School of Pharmacy, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Raquel Herrero
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Natalia Torroglosa
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Alexandra Pumarola
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Narine Fischer-Albiol
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Sofia Campos-Moreno
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Sofia Sabaté
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Àngels Alcina
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Sandra Mancilla
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain
| | - Belén García
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain
| | - Monserrat Llaguno-Munive
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando 22, 14080 Ciudad de México, CDMX, Mexico
| | - Zamira V Díaz-Riascos
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain
| | - Cláudia Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal
| | - Simó Schwartz
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Roser Ferrer-Costa
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Ibane Abasolo
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain; Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Pilar Sánchez-Gómez
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, Instituto de Salud Carlos III, Ctra. Pozuelo-Majadahonda, km 2, 28220 Madrid, Spain
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IUCS-CESPU, Rua Central de Gandra 1317, Gandra 4585-116, Portugal
| | - Diana Rafael
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain.
| | - Fernanda Andrade
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Department of Pharmacy and Pharmaceutical Technology and Physicochemistry, Faculty of Pharmacy and Food Sciences, School of Pharmacy, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain.
| |
Collapse
|
5
|
Zhao C, Guo Y, Chen Y, Shang G, Song D, Wang J, Yang J, Zhang H. RETRACTED: Zinc finger Protein207 orchestrates glioma migration through regulation of epithelial-mesenchymal transition. ENVIRONMENTAL TOXICOLOGY 2025; 40:E59-E73. [PMID: 38591780 DOI: 10.1002/tox.24271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Glioma represents the predominant primary malignant brain tumor. For several years, molecular profiling has been instrumental in the management and therapeutic stratification of glioma, providing a deeper understanding of its biological complexity. Accumulating evidence unveils the putative involvement of zinc finger proteins (ZNFs) in cancer. This study aimed to elucidate the role and significance of ZNF207 in glioma. METHODS Utilizing online data such as The Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA), the Genotype-Tissue Expression (GTEx) project, the Clinical Proteomic Tumor Analysis Consortium (CPTAC), and the Human Protein Atlas (HPA) databases, in conjunction with bioinformatics methodologies including GO, KEGG, GSEA, CIBERSORT immune cell infiltration estimation, and protein-protein interaction (PPI) analysis, enabled a comprehensive exploration of ZNF207's involvement in gliomagenesis. Immunohistochemistry and RT-PCR techniques were employed to validate the expression level of ZNF207 in glioma samples. Subsequently, the biological effects of ZNF207 on glioma cells were explored through in vitro assays. RESULTS Our results demonstrate elevated expression of ZNF207 in gliomas, correlating with unfavorable patient outcomes. Stratification analyses were used to delineate the prognostic efficacy of ZNF207 in glioma with different clinicopathological characteristics. Immunocorrelation analysis revealed a significant association between ZNF207 expression and the infiltration levels of T helper cells, macrophages, and natural killer (NK) cells. Utilizing ZNF207 expression and clinical features, we constructed an OS prediction model and displayed well discrimination with a C-index of 0.861. Moreover, the strategic silencing of ZNF207 attenuated glioma cell advancement, evidenced by diminished cellular proliferation, weakened cell tumorigenesis, augmented apoptotic activity, and curtailed migratory capacity alongside the inhibition of the epithelial-mesenchymal transition (EMT) pathway. CONCLUSIONS ZNF207 may identify as a prospective biomarker and therapeutic candidate for glioma prevention, providing valuable insights into understanding glioma pathogenesis and treatment strategies.
Collapse
Affiliation(s)
- Chao Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yuduo Guo
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yujia Chen
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Guanjie Shang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Dixiang Song
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jun Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jingjing Yang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Hongwei Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Jiang S, Zhu L, Jiang S. Phosducin inhibits the cell proliferation and promotes the antitumor effect of temozolomide in glioma. Biochem Pharmacol 2025; 235:116841. [PMID: 40024352 DOI: 10.1016/j.bcp.2025.116841] [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: 10/23/2024] [Revised: 02/02/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Malignant gliomas are the most lethal form of brain cancer, characterized by rapid cell growth, substantial molecular heterogeneity, and a propensity for invasion into critical brain regions. Phosducin (PDC) is recognized for its involvement in sensory signal transmission, blood pressure regulation, and thyroid gland endocrine functions. However, the role of PDC in cell proliferation, drug sensitization, and its connection to RNA m6A modification in gliomas remains unclear. In this study, RNA sequencing analysis was performed on U251 glioma cells with knockdown and overexpression of fat mass and obesity-associated protein (FTO). The results revealed that FTO negatively regulates PDC expression. This finding was corroborated in U87, U251, and A172 glioma cells via qRT-PCR and western blot analysis. Additionally, MTT and EdU assays revealed that PDC overexpression inhibited cell proliferation, while PDC knockdown accelerated it. Moreover, the proliferation-enhancing effect of FTO overexpression was reduced by PDC overexpression, and the proliferation-inhibiting effect of FTO knockdown was reversed by PDC knockdown. These findings suggest that PDC serves as a functional target of FTO. Furthermore, PDC enhanced the antitumor efficacy of temozolomide (TMZ). In summary, this study demonstrates for the first time that PDC plays a crucial role in regulating cell proliferation and TMZ sensitivity in glioma cells, providing a potential therapeutic target to improve treatment outcomes for the patients with glioma.
Collapse
Affiliation(s)
- Shibin Jiang
- Department of Biology, School of Life Science, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Lifang Zhu
- Department of Biology, School of Life Science, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Songshan Jiang
- Department of Biology, School of Life Science, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| |
Collapse
|
7
|
Ye X, Zhong H, Liu L, Huang J, Xia Z, Tang Z, Wei W, Huang W, Ye Y, Jiang Q. A novel and high-performance tumor inhibitor of La, N co-doped carbon dots for U251 and LN229 cells. Colloids Surf B Biointerfaces 2025; 249:114520. [PMID: 39823950 DOI: 10.1016/j.colsurfb.2025.114520] [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: 11/19/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
To address the medical challenges posed by glioblastoma, a novel and high-performance tumor inhibitor (La@FA-CDs) composed of folic acid and lanthanum nitrate hexahydrate, was successfully synthesized and demonstrated effectiveness in inhibiting the growth of U251 and LN299 cells. The microstructure of La@FA-CDs was extensively analyzed by FTIR, UV-Vis, XPS, TEM, AFM NMR, and nanoparticle size analyzer. The optical and electrical properties of La@FA-CDs were characterized using a fluorescence spectrometer and a zeta potential analyzer. Biological assays, including the CCK8 proliferation assay, scratch assay, flow cytometry, cytoskeleton staining, and live/dead staining were conducted to assess antitumor properties and cytotoxicity. The result revealed that the La50 %@FA-CDs demonstrated significantly enhanced antitumor activity relative to the undoped sample. Furthermore, the La50 %@FA-CDs demonstrated a dose-dependent cytotoxic effect on two glioblastoma cell lines U251 and LN299. The findings of this study suggested that treatment with La50 %@FA-CDs effectively inhibited migration and proliferation while promoting apoptosis in glioblastoma cells. Meanwhile, the La50 %@FA-CDs showed minimal cytotoxic effects on HEK 293 and HUVEC cells under standard conditions, with only slight toxicity observed in HUVEC cells at high (500 µM) concentrations. These results suggest that La50 %@FA-CDs could be a promising therapeutic agent for glioblastoma treatment, demonstrating both effective inhibition and favorable safety profiles.
Collapse
Affiliation(s)
- Xinyun Ye
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China
| | - Huanglian Zhong
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China
| | - Lin Liu
- School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi Province 341000, China
| | - Jingtao Huang
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China
| | - Zhuquan Xia
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China
| | - Zhiji Tang
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China
| | - Wenjin Wei
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China
| | - Weilong Huang
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China.
| | - Yuwei Ye
- Faculty of Materials Metallurgy and Chemistry, Jiangxi University of Science and Technology, Ganzhou 341000, China.
| | - Qiuhua Jiang
- Department of Neurosurgery, The Affiliated Ganzhou Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China.
| |
Collapse
|
8
|
Wang C, Xiong X, Li C, Lu Y, Zhang Y, Tian H, Xu C, Ma T, Wang J, Zhang J, Wang L. Sophoricoside Inhibited Glioblastoma Cell Progression Through Activated AMP-Activated Protein Kinase (AMPK). Mol Carcinog 2025; 64:816-828. [PMID: 39878044 DOI: 10.1002/mc.23889] [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: 07/05/2024] [Revised: 12/06/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, with a mean survival of less than 2 years. Unique brain structures and the microenvironment, including blood-brain barriers, put great challenges on clinical drug development. Sophoricoside (Sop), an isoflavone glycoside isolated from seeds of Sophora japonica L., is one of the active constituents of traditional Chinese medicine and found to inhibit the bioactivity of cytokines (e.g., interleukin-5) and inflammatory responses, as well as to attenuate glucose and lipid metabolism in related diseases. However, the effects of Sop on cancer progression have not been systemically investigated. In this study, we performed a comprehensive investigation of Sop's function in GBM using colony formation and Transwell assays in vitro, along with subcutaneous xenograft tumor analysis in vivo. We employed RNA sequencing and bioinformatics analysis in conjunction with Western blotting (WB) and reverse transcription-quantitative polymerase chain reaction (RT-PCR) to explore the underlying mechanism. Our results demonstrated that Sop suppressed U251 cell proliferation and metastasis in vitro and inhibited the tumorigenic behavior of U251 cells in vivo. Further investigations revealed a positive correlation between the levels of activated AMP-activated protein kinase (AMPK) and Sop treatment; notably the application of the AMPK inhibitor, compound C (CC), abolished inhibitory effects of Sop on the malignant phenotype of U251 cells. These findings suggest the potential application of Sop in GBM treatment and highlight opportunities for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Changquan Wang
- Department of Neurology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Xuehui Xiong
- Department of Neurosurgery, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Chang Li
- Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuan Lu
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Yan Zhang
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Han Tian
- Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chunlin Xu
- Department of Neurology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Tengfei Ma
- Huanggang Institute of Translational Medicine, Huanggang, China
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Jinhua Wang
- Department of Neurology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Jianqing Zhang
- Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Neurosurgery, Huanggang Central Hospital of Yangtze University, Huanggang, China
| |
Collapse
|
9
|
Nåhls NS, Anttonen A, Nuutinen M, Saarto T, Carpén T. The impact of palliative care contact on the use of hospital resources at the end of life for brain tumor patients; a nationwide register-based cohort study. J Neurooncol 2025; 172:549-556. [PMID: 39833624 PMCID: PMC11968532 DOI: 10.1007/s11060-025-04939-9] [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: 12/12/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE The aim of this nationwide retrospective cohort study was to evaluate the timing of the first specialist palliative care (SPC) contact and its impact on the use of hospital resources at the end of life in patients with brain tumors. MATERIALS AND METHODS The analysis comprised 373 brain tumor patients who died during 2019 in Finland. Patients were divided into two groups according to the time of first SPC contact: early, i.e. first SPC contact more than 30 days before death, and late, i.e. no SPC contact or 30 days or less before death. RESULTS 216 (58%) were male, with a mean age of 67 years (range 18-94). SPC contact was established for 102 (27%) patients and the median time of first SPC contact before death was 76 days. Patients with an early SPC contact had fewer outpatient clinic contacts (28% vs. 53%; p-value < 0.001) and fewer hospitalization (10% vs. 37%; p-value < 0.001) in secondary care compared with patients with late SPC contact. Early SPC contact had no impact on emergency department contacts. Patients with early SPC contact were more likely to die at long term care facility or in SPC wards instead of hospital (p-value < 0.001) compared to patients with late SPC contact (hospital deaths 51% vs. 80%, respectively). CONCLUSIONS Early SPC contact reduced the burden on secondary care for brain tumor patients in the last months of life. Palliative care contact should be offered early to all brain tumor patients.
Collapse
Affiliation(s)
- Nelli-Sofia Nåhls
- Department of Oncology, Vaasa Central Hospital, The Wellbeing Services County of Ostrobothnia, Vaasa, Finland.
- Department of Oncology, Comprehensive Cancer Centre, University of Helsinki, Helsinki, Finland.
| | - Anu Anttonen
- Department of Radiotherapy, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | | | - Tiina Saarto
- Department of Palliative Care, Comprehensive Cancer Center, Helsinki University Hospital, and Faculty of Medicine, Helsinki University, Helsinki, Finland
| | - Timo Carpén
- Department of Palliative Care, Comprehensive Cancer Center, Helsinki University Hospital, and Faculty of Medicine, Helsinki University, Helsinki, Finland
| |
Collapse
|
10
|
Chen Y, Zhang J, Xu H. Recent developments in cuproptosis of glioblastoma. Pathol Res Pract 2025; 269:155939. [PMID: 40164043 DOI: 10.1016/j.prp.2025.155939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Glioblastoma (GBM) is the most malignant tumor within the central nervous system, attributed to its high-grade malignancy, propensity for recurrence, refractoriness to conventional therapeutic modalities, and the suboptimal efficacy of current targeted therapies. Hence, there is an urgent need to identify more efficacious molecular targets for the therapeutic intervention of GBM. The regulated cell death (RCD) has specific signaling factors and signaling pathways. Hence, targeting RCD is considered to be one of the effective targeted therapies for GBM. At present, cuproptosis is a novel form of RCD, characterized by a distinct molecular mechanism that differentiates it from apoptosis, pyroptosis, necroptosis, and ferroptosis. It is characterized by its principal mechanisms, which include copper dependency, the accumulation of acylated proteins, and the reduction of Fe-S cluster-containing proteins. These processes collectively induce proteotoxic stress, culminating in cell death. In previous studies, copper-ionized formulations have demonstrated cytotoxic effects on gliomas. Thus, the key factors of cuproptosis may be able to serve as a new target for GBM treatment. This review delves into several pivotal aspects, including the discovery of cuproptosis, the impact of copper homeostasis on tumorigenesis, the role of cuproptosis in GBM, and its potential as a therapeutic target in molecular targeted therapy for GBM. Hence, this article could reveal novel strategies for GBM treatment.
Collapse
Affiliation(s)
- Yajia Chen
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Jingxian Zhang
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Hongwu Xu
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Wandao Rd, 78, Wanjiang Subdistrict, Dongguan, Guangdong Province 523059, China; Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China.
| |
Collapse
|
11
|
Zhan Y, Hao Y, Wang X, Guo D. Advances of artificial intelligence in clinical application and scientific research of neuro-oncology: Current knowledge and future perspectives. Crit Rev Oncol Hematol 2025; 209:104682. [PMID: 40032186 DOI: 10.1016/j.critrevonc.2025.104682] [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: 11/01/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
Brain tumors refer to the abnormal growths that occur within the brain's tissue, comprising both primary neoplasms and metastatic lesions. Timely detection, precise staging, suitable treatment, and standardized management are of significant clinical importance for extending the survival rates of brain tumor patients. Artificial intelligence (AI), a discipline within computer science, is leveraging its robust capacity for information identification and combination to revolutionize traditional paradigms of oncology care, offering substantial potential for precision medicine. This article provides an overview of the current applications of AI in brain tumors, encompassing the primary AI technologies, their working mechanisms and working workflow, the contributions of AI to brain tumor diagnosis and treatment, as well as the role of AI in brain tumor scientific research, particularly in drug innovation and revealing tumor microenvironment. Finally, the paper addresses the existing challenges, potential solutions, and the future application prospects. This review aims to enhance our understanding of the application of AI in brain tumors and provide valuable insights for forthcoming clinical applications and scientific inquiries.
Collapse
Affiliation(s)
- Yankun Zhan
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Yanying Hao
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Xiang Wang
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China.
| | - Duancheng Guo
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
12
|
Aroujo J, Parker H, Boari A, Mason E, Otakpor MU, Betancourt T, Kornienko A, Ciavatta ML, Carbone M, Evidente A, Taube JH, Romo D. Derivatization of ophiobolin A and cytotoxicity toward breast and glioblastoma cancer stem cells: Varying the ketone and unsaturated aldehyde moieties. Bioorg Med Chem Lett 2025; 120:130112. [PMID: 39880174 DOI: 10.1016/j.bmcl.2025.130112] [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: 11/22/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
To gain further insights into the importance of the unsaturated 1,4-ketoaldehyde moiety of ophiobolin A (OpA) for the potency and selectivity observed toward cancer stem cells, several derivatives were synthesized through controlled reduction and oxidations of the unsaturated aldehyde and ketone moieties. Structure elucidation of these new OpA derivatives was achieved through detailed NMR studies and comparison to OpA and known isolated congeners possessing variations in these regions. The relative stereochemistry of the newly generated stereocenters was determined by coupling constants in conjunction with conformational analyses (DFT) of the synthetic derivatives. The cytotoxicity of these derivatives was studied against breast cancer and glioblastoma cell lines possessing stem-cell like properties. In addition, the comparative activity toward HMLE and HMLE-TWIST mammary epithelial cells was studied, with the latter cell line representing an epithelial mesenchymal transition positive (EMT+) cell line.
Collapse
Affiliation(s)
- Jaquelin Aroujo
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States
| | - Haleigh Parker
- Department of Biology, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States
| | - Angela Boari
- Institute of Sciences of Food Production, National Research Council, Via Amendola 122/O, 70125 Bari, Italy
| | - Evan Mason
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States
| | - Mackenzie U Otakpor
- Department of Chemistry and Biochemistry, Texas State University, 601 University Dr., TX 78666, United States
| | - Tania Betancourt
- Department of Chemistry and Biochemistry, Texas State University, 601 University Dr., TX 78666, United States
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, 601 University Dr., TX 78666, United States.
| | - Maria Letizia Ciavatta
- Institute of Biomolecular Chemistry, National Research Council (CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Marianna Carbone
- Institute of Biomolecular Chemistry, National Research Council (CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Antonio Evidente
- Institute of Biomolecular Chemistry, National Research Council (CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy.
| | - Joseph H Taube
- Department of Biology, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States.
| | - Daniel Romo
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States.
| |
Collapse
|
13
|
Wang M, Sun G, Fan Y, Sima G, Sun X, Qiu T, Li X. Hydroxymethyltransferase 2 promotes the development of glioblastoma by mediating WTAP regulation of PTEN N6-methyladenosine modification. Metab Brain Dis 2025; 40:195. [PMID: 40299174 DOI: 10.1007/s11011-025-01621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
Characterized by rapid proliferation and therapeutic resistance, glioblastoma (GBM) represents the deadliest primary CNS neoplasm, demonstrating a low survival rate and high mortality rate in patients. This is mainly related to the development of GBM more specifically due to the abnormal metabolism within cells. SHMT2 (serine hydroxymethyltransferase 2) acts as a pivotal metabolic regulator in neoplastic cells, driving one-carbon unit transfer essential for nucleotide biosynthesis. Here, we explored the mechanism of SHMT2 mediated GBM occurrence. In this study, SHMT2 expression was assessed in GBM cells and tissues. In vitro experiments were performed to investigate the functional role of SHMT2. The detailed mechanisms of SHMT2-mediated cell metabolism were addressed. Xenograft model analysis explored the influence of SHMT2 on GBM development. The expression level of SHMT2 in GBM clinical tissues and cell lines is higher than that in normal tissues. The downregulation of SHMT2 inhibits the proliferation ability and metabolic process of GBM cell lines. Mechanism dissection revealed that SHMT2 enhanced phosphatase and tensin homolog (PTEN) N6-methyladenosine (m6A) modification through the endogenous methyl donor SAM mediated by SHMT2 via serine/glycine one carbon metabolic networks. In addition, Xenograft model analysis showed that knockdown of SHMT2 inhibited the development of GBM tumors. SHMT2 promotes the tumorigenesis of glioblastoma by regulating the m6A modification of PTEN.
Collapse
Affiliation(s)
- Meng Wang
- Department of Neurosurgery, First Affiliated Hospital, Soochow University, No. 899 Pinghai Road, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Guangwei Sun
- Department of Neurosurgery, Danyang People's Hospital, No.2 Xinmin West Road, Danyang, Zhenjiang, 212300, Jiangsu, China
| | - Yongzhong Fan
- Department of Neurosurgery, Danyang People's Hospital, No.2 Xinmin West Road, Danyang, Zhenjiang, 212300, Jiangsu, China
| | - Guozhong Sima
- Department of Neurosurgery, Danyang People's Hospital, No.2 Xinmin West Road, Danyang, Zhenjiang, 212300, Jiangsu, China
| | - Xifeng Sun
- Department of Neurosurgery, Danyang People's Hospital, No.2 Xinmin West Road, Danyang, Zhenjiang, 212300, Jiangsu, China
| | - Tao Qiu
- Department of Neurosurgery, Danyang People's Hospital, No.2 Xinmin West Road, Danyang, Zhenjiang, 212300, Jiangsu, China
| | - Xiangdong Li
- Department of Neurosurgery, First Affiliated Hospital, Soochow University, No. 899 Pinghai Road, Gusu District, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
14
|
Chen P, Liu Y, Huang H, Li M, Xie H, Roy S, Gu J, Jin J, Deng K, Du L, Guo B. Genetically Engineered IL12/CSF1R-Macrophage Membrane-Liposome Hybrid Nanovesicles for NIR-II Fluorescence Imaging-Guided and Membrane-Targeted Mild Photothermal-Immunotherapy of Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500131. [PMID: 40279543 DOI: 10.1002/advs.202500131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/19/2025] [Indexed: 04/27/2025]
Abstract
It is a big challenge for precision therapy of glioblastoma, mainly due to the existence of blood-brain barrier (BBB), tumor immunosuppressive microenvironment (TIM), and lack of efficient treatment paradigms. Herein, a theranostic nanoplatform for the second near-infrared window (NIR-II) fluorescence imaging-guided membrane-targeted mild photothermal-immunotherapy of glioblastoma using genetically engineered CSF1R/IL12-macrophage membrane (MM)-liposome hybrid nanovesicles, is reported. By mimicking lipophilic membrane probe (Dil) with octadecyl chains, a NIR-II emissive photothermal dye (IRC18), which realizes labeling of nanovesicle lipid bilayers for biodistribution tracing, glioblastoma diagnosis, and molecular imaging of tumoral microenvironment, is synthesized. Importantly, MM and c-RGD-decorated liposome together offer BBB crossing, tumor targeting, and long-term circulation; while, the genetically overexpressed CSF1R and IL12 on MM surface contribute to effective modulation of M2-to-M1 macrophage repolarization and local promotion of T cell cytotoxicity in glioblastoma microenvironment, respectively. Notably, through membrane fusion, IRC18 dyes translocate from nanovesicle lipid bilayers to glioblastoma membranes, which achieve membrane-targeted mild photothermal therapy to ablate primary tumor and induce immunogenic cell death to promote antigen presentation. More importantly, the combined blockade of the CSF1-CSF1R axis and IL-12 enrichment not only reprograms the tumor microenvironment through macrophage M1 repolarization but also activates cytotoxic T cells, ultimately achieving complete glioblastoma eradication. This research provides an efficient theranostic paradigm for glioblastoma treatment.
Collapse
Affiliation(s)
- Pengfei Chen
- Department of Traumatic Orthopedics, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Yue Liu
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Haiyan Huang
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Menglong Li
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Hui Xie
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jian Jin
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Kai Deng
- Department of Traumatic Orthopedics, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Lixin Du
- Department of Medical Imaging, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| |
Collapse
|
15
|
Yi GZ, Lin JF, Lu YX, Li ZY, Qu SQ, Feng WY, Huang GL. Clinical, pathological, radiological features and prognosis of epithelioid glioblastoma: a retrospective single center study. J Neurooncol 2025:10.1007/s11060-025-05046-5. [PMID: 40266499 DOI: 10.1007/s11060-025-05046-5] [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: 01/13/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Epithelioid glioblastoma (eGBM) is a rare variant of glioblastoma (GBM). Due to the limited number of reported cases, our understanding of eGBM remains constrained. The updated definition of GBM has further highlighted the need for more comprehensive studies on this rare subtype. MATERIALS AND METHODS This study retrospectively reviewed 17 cases of eGBM diagnosed at a single center between 2016 and 2023. Detailed clinicopathological data were analyzed, including clinical presentation, pathological characteristics, radiological findings, treatment modalities, and patient outcomes. RESULTS The cohort comprised 8 females and 9 males with an average age of 40.88 years. Tumors were predominantly located in the supratentorial region (64.71%, 11/17), while uncommon locations included the basal ganglia (11.76%, 2/17), sellar region (5.88%, 1/17), and pineal region (5.88%, 1/17). Multi-modal MRI scans revealed consistent features such as T1 contrast enhancement, elevated Cho/NAA ratios, increased nCBF and nCBV values, and disruption of white matter tracts. Additional radiological findings included mild peritumoral edema (64.71%, 11/17), hemorrhage (17.65%, 3/17), dural tail sign (35.29%, 6/17), and necrosis (29.41%, 5/17). Pathologically, all cases exhibited microvascular proliferation, IDH1 wild-type status, and high Ki-67 indices. Other notable findings included EMA positivity or partial positivity (47.06%, 8/17), INI-1 positivity (100%, 11/11), BRAF-V600E mutation (76.47%, 13/17), necrosis (82.35%, 14/17), and leptomeningeal dissemination (29.41%, 5/17). All patients underwent gross total resection (GTR), and 14 received adjuvant chemoradiotherapy. The median overall survival was 12.53 months. Two patients who received TTFields therapy survived for 12.53 and 16.63 months, respectively. Two patients treated with BRAF-V600E-targeted therapy had survival times of 16.93 and 22.67 months, respectively. CONCLUSION eGBM is a distinct and aggressive variant of GBM characterized by shorter survival times. The variability in MRI features and unusual tumor locations can lead to misdiagnosis, which multi-modal MRI may help mitigate. High frequencies of BRAF V600E mutations and INI-1 positive expression are observed in eGBM. Combined therapies, including TTFields and BRAF V600E-targeted treatments, appear to be associated with improved outcomes.
Collapse
Affiliation(s)
- Guo-Zhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University Guangzhou, Dadao Bei Street Guangzhou, Guangdong, 510515, People's Republic of China
| | - Jin-Feng Lin
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University Guangzhou, Dadao Bei Street Guangzhou, Guangdong, 510515, People's Republic of China
| | - Yan-Xia Lu
- Department of Pathology, Nanfang Hospital, Southern Medical University Guangzhou, Guangdong, 510515, People's Republic of China
| | - Zhi-Yong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University Guangzhou, Dadao Bei Street Guangzhou, Guangdong, 510515, People's Republic of China
| | - Shan-Qiang Qu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University Guangzhou, Dadao Bei Street Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wen-Yan Feng
- Department of Clinical Medicine, Nanfang Hospital, Southern Medical University Guangzhou, Dadao Bei Street, Guangdong, 510515, People's Republic of China.
| | - Guang-Long Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University Guangzhou, Dadao Bei Street Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
16
|
Shang Y, Liang Y, Zhang B, Wu W, Peng Y, Wang J, Zhang M, Niu C. Periostin-mediated activation of NF-κB signaling promotes tumor progression and chemoresistance in glioblastoma. Sci Rep 2025; 15:13955. [PMID: 40263417 PMCID: PMC12015317 DOI: 10.1038/s41598-025-92969-8] [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] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive form of diffuse glioma, characterized by high lethality. Temozolomide (TMZ)-based chemotherapy is a standard treatment for GBM, but development of chemoresistance poses a significant therapeutic challenge. Despite advances in understanding GBM biology, the mechanisms driving TMZ resistance remain unclear. Identifying vital molecular players involved in this resistance is crucial for developing new therapies. Our results indicated that periostin (POSTN) was significantly upregulated in GBM cell lines and patient samples, correlating with poorer clinical outcomes. POSTN overexpression enhanced GBM cell proliferation, migration, invasion, and chemoresistance, while lentiviral suppression of POSTN significantly reduced these behaviors. In vivo, bioluminescence imaging further confirmed the enhanced tumor growth associated with POSTN overexpression. Bioinformatics analysis was performed to explore the underlying molecular mechanism. The results revealed a strong correlation between POSTN and epithelial-mesenchymal transition (EMT) process and the tumor necrosis factor α (TNFα)-NF-κB signaling pathway. Moreover, exogenous POSTN silencing reduced IκB-kinase α (IKKα) phosphorylation, thereby decreasing NF-κB expression by limiting IκBα degradation. Collectively, our study demonstrated that POSTN-induced activation of NF-κB signaling and EMT processes promoted the malignancy and chemoresistance of GBM, suggesting that POSTN may serve as a reliable prognostic biomarker and potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Yu Shang
- PET-CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
- Future Technology Institute, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Yuxia Liang
- Department of Physical Examination, The First Hospital Affiliated to Xi'an Jiao Tong University, Xi'an, 710061, Shaanxi, China
| | - Beichen Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yihao Peng
- Future Technology Institute, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Jin Wang
- Future Technology Institute, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Ming Zhang
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Chen Niu
- PET-CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
- Future Technology Institute, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
- Department of Information, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
17
|
Hu Z, Li W, Wei L, Ma J. Lactoferrin in cancer: Focus on mechanisms and translational medicine. Biochim Biophys Acta Rev Cancer 2025; 1880:189330. [PMID: 40274081 DOI: 10.1016/j.bbcan.2025.189330] [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: 12/05/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Lactoferrin is an iron-binding glycoprotein that provides natural protective effects to the human body. Its biological properties, including antibacterial, antiviral, anti-inflammatory, immune-regulatory, and iron metabolism-regulating functions, have been extensively studied. With further research, lactoferrin's impact on tumorigenesis and tumor microenvironment has become increasingly evident, as it inhibits tumor proliferation, invasion, and metastasis through multiple pathways. This article summarizes the molecular mechanisms underlying lactoferrin's anticancer effects, explores its association with the malignant progression of various cancers, and highlights its clinical translational potential as a potential cancer biomarker and drug delivery carrier to enhance anticancer therapy efficiency. Due to the high safety profile of lactoferrin, its widespread application in the field of cancer treatment is highly anticipated.
Collapse
Affiliation(s)
- Zhengyu Hu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Wenchao Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Lingyu Wei
- Laboratory of Clinical Research Center, Department of Pathology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China.
| |
Collapse
|
18
|
Bardhan M, Muneer MA, Khare A, Minesh Shah R, Kaur A, Vasipalli SS, Suresh V, Podder V, Ahluwalia M, Odia Y, Chen Z. Advances in stem cell-based therapeutic transfers for glioblastoma treatment. Expert Rev Neurother 2025:1-17. [PMID: 40245098 DOI: 10.1080/14737175.2025.2490543] [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: 09/23/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Glioblastoma (GBM), a highly malignant brain tumor, has a poor prognosis despite standard treatments like surgery, chemotherapy, and radiation. Glioblastoma stem cells (GSCs) play a critical role in recurrence and therapy resistance. Stem cell-based therapies have emerged as innovative approaches, leveraging the tumor-targeting abilities of stem cells to deliver treatments directly to GBM. AREAS COVERED This review focuses on using intact stem cells or subtypes for GBM therapy, excluding antigenic characteristics. The stem cell-based therapies explored include neural, mesenchymal, glioblastoma, hematopoietic and adipose-derived stem cells that have been investigated in both clinical and preclinical settings. A systematic search in PubMed, EMBASE, ClinicalTrials.gov, and Scopus had identified research up until January 2024. Key mechanisms reviewed include immune modulation, angiogenesis inhibition, and apoptosis induction. Discussion of completed and ongoing trials include emphasis on safety, efficacy, challenges, and study design limitations. EXPERT OPINION Stem cell-based therapies hold promise for treating GBM by targeting GSCs and improving treatment outcomes. Despite some potential advantages, challenges such as tumorigenesis risks, delivery complexities, and sustained therapeutic effects persist. Future research should prioritize optimizing stem cell modifications, combining them with current treatments, and conducting large-scale trials to ensure safety and efficacy. Integrating stem cell therapies into GBM treatment could provide more effective and less invasive options for patients.
Collapse
Affiliation(s)
- Mainak Bardhan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | | - Abhinav Khare
- All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | | | - Anmol Kaur
- Lady Hardinge Medical College, New Delhi, India
| | - Sonit Sai Vasipalli
- Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Vinay Suresh
- King George's Medical University, Lucknow, India
| | - Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Manmeet Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Yazmin Odia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Zhijian Chen
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| |
Collapse
|
19
|
Gadhave D, Quadros M, Ravula S, Ugale AR, Alkyam M, Perron JC, Gupta V. Quality by design enabled development & in-vitro assessment of a Nanoemulgel formulation for Nose-to-Brain delivery of Nintedanib for glioblastoma multiforme treatment. Int J Pharm 2025; 676:125632. [PMID: 40268208 DOI: 10.1016/j.ijpharm.2025.125632] [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: 01/11/2025] [Revised: 04/20/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
Glioblastoma multiforme (GBM) is a deadly malignant brain tumor that spreads uncontrollably and invades the surrounding brain parenchyma. GBM treatment remains challenging due to the rigid blood-brain barrier, limiting therapeutic entry into the brain. Therefore, the current study focused on formulating a Nintedanib (Nint) loaded in-situ Nanoemulgel (Nint-Nanoemulgel) and exploring its permeation and therapeutic potential under in-vitro models to address these limitations. Nint-Nanoemulgel was optimized through the QbD-enabled Box-Behnken design. Optimized Nint-Nanoemulgel revealed significant globule size (27.4 ± 0.8 nm), PDI (0.17 ± 0.01), % encapsulation efficiency (93.5 ± 3.5 %), zeta potential (-4.7 ± 0.6 mV), %T (98.2 ± 0.2 %), pH (6.0 ± 0.2), and viscosity (2.59 ± 0.24 cP) at 25 °C. A cumulativein-vitro release study revealed 87.4 ± 1.9 % Nint release through Nanoemulgel after 12 h while 90.1 ± 2.1 % release after 24 h.The cytotoxicity potential of developed Nint-Nanoemulgel was screened in GBM cell lines, demonstrating a > 2-fold reduction in IC50 than plain Nint. However, after treatment with 100 µM of Nint-Nanoemulgel, % growth inhibition was found to be 91.0 ± 1.0 %, 92.1 ± 1.3 %, and 82.7 ± 1.0 % in LN229, U87, and U138 cell lines, respectively. Further,in-vitro cellular uptake exhibited significant coumarin cellular internalization through nanoformulations against coumarin solution. Moreover, clonogenic and scratch assay studies demonstrated the ability of Nint-NE to inhibit cell proliferation and colony formation. However, the outcomes of the live-dead assay demonstrated more cell death in Nint-nanoformulation-treated spheroids than in Nint-treated spheroids. Nint-Nanoemulgel improved intracellular permeation and demonstrated a 2-fold increase in Nint transport across the RPMI-2650 epithelial monolayer. Finally, favorable outcomes of intranasal Nint-Nanoemulgel could provide a novel avenue for the safe and effective delivery of Nint in GBM patients.
Collapse
Affiliation(s)
- Dnyandev Gadhave
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Mural Quadros
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Sravani Ravula
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Akanksha R Ugale
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Mayssam Alkyam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Jeanette C Perron
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA.
| |
Collapse
|
20
|
Wang F, Chen Y, Huang R, Lu D, Zhang J, Yang Y, Dang H, Liu M, Chen Z, Sun X, Wang Z. Identification of SURF4 and RALGAPA1 as promising therapeutic targets in glioblastoma and pan-cancer through integrative multi-omics, CRISPR-Cas9 screening and prognostic meta-analysis. Cancer Immunol Immunother 2025; 74:175. [PMID: 40249536 PMCID: PMC12008100 DOI: 10.1007/s00262-025-04034-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: 01/15/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025]
Abstract
Glioblastoma (GBM) is the most aggressive and malignant type of primary brain tumor, with a median survival time of less than two years and a uniformly poor prognosis, despite multimodal therapeutic approaches, which highlights an urgent need for novel therapeutic targets. In this study, by integrative multi-omics analysis from CPTAC database, DepMap database and seven independent GBM cohorts, four hub genes (CD44, SURF4, IGSF3 and RALGAPA1) were identified as essential genes regulated by cancer driver genes with robust prognostic value. GBM multi-omics data from public and in-house cohorts validated that CD44 and SURF4 might be synthetic lethal partners of loss-of-function tumor suppressor genes. Analysis for immune-related pathway activity revealed complex regulation relationships of the four hub genes in tumor microenvironment (TME). Further investigation on SURF4 in pathway activity, immune therapy response and drug sensitivity proposed that SURF4 emerged as a promising therapeutic target for GBM, even for pan-cancer. Pan-cancer multi-omics exploration suggested that RALGAPA1 may be a tumor suppressor gene. By screening the first-generation and second-generation DepMap database, four genes (CCDC106, GAL3ST1, GDI2 and HSF1) might be considered as synthetic targets after mutation of RALGAPA1 as a tumor suppressor gene.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Yuxuan Chen
- Suzhou Medical College of Soochow University, Suzhou, 215127, Jiangsu Province, China
| | - Run Huang
- Suzhou Medical College of Soochow University, Suzhou, 215127, Jiangsu Province, China
| | - Dengfeng Lu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Juyi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Yanbo Yang
- Department of Oncology, Division of Pediatric Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hanhan Dang
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100000, China
| | - Meirong Liu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Zhouqing Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China.
| | - Xiaoou Sun
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
21
|
YANG MING, CHU LIANGZHAO, LIN SHUKAI, PENG HAN, LONG NIYA, XU KAYA, YANG HUA, HAN FENG, LIU JIAN. The alternatively spliced diacylglycerol kinase gamma-Δ exon13 transcript generated under hypoxia promotes glioblastoma progression. Oncol Res 2025; 33:1189-1198. [PMID: 40296900 PMCID: PMC12034013 DOI: 10.32604/or.2024.055102] [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: 06/17/2024] [Accepted: 11/13/2024] [Indexed: 04/30/2025] Open
Abstract
Background Glioblastoma (GBM) is one of the most malignant types of central nervous system tumors. Oxygen deprivation in the tumor microenvironment is thought to be an important factor in promoting GBM progression. However, the mechanisms of hypoxia-promoted tumor progression remain elusive. Methods Alternative splicing of diacylglycerol kinase gamma (DGKG)-Δ exon13 was amplified and verified by PCR-Sanger sequencing. The functions of DGKG and DGKG-Δ exon13 were analyzed by Cell counting kit-8 (CCK-8), Transwell, Matrigel-transwell experiments, and in vivo orthotropic GBM animal models. Transcriptome analyses were done to find out the regulated genes. Results In this study, we found that a new transcript DGKG-Δ exon13 was generated in GBM under hypoxia via alternative splicing. Moreover, the results of CCK-8, Transwell, and Matrigel-transwell experiments showed that the proliferation, migration, and invasion abilities of U87-MG and T98G were decreased after DGKG knockdown. Compared to wild-type DGKG, DGKG-Δ exon13 overexpression significantly promoted cellular proliferation, migration, and invasion abilities in GBM. Furthermore, in vivo, orthotropic GBM animal models analysis showed that the tumor volumes were much smaller in the DGKG knockdown group. However, the tumor sizes in the DGKG and DGKG-Δ exon13 rescue groups were restored, especially in the DGKG-Δ exon13 group. Transcriptome analysis revealed that MORC1, KLHDC7B, ATP1A2, INHBE, TMEM119, and FGD3 were altered significantly when DGKG was knocked down. IL-16, CCN2, and EFNB3 were specifically regulated by DGKG-Δ exon13. Conclusions Our study found that hypoxia-induced alternative splicing transcript DGKG-Δ exon13 promotes GBM proliferation and infiltration, which might provide a new potential target for the clinical treatment and diagnosis of GBM.
Collapse
Affiliation(s)
- MING YANG
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - LIANGZHAO CHU
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - SHUKAI LIN
- Department of Neurosurgery, Sanya Central Hospital (Hainan Third People’s Hospital), Sanya, 572000, China
| | - HAN PENG
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - NIYA LONG
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - KAYA XU
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - HUA YANG
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - FENG HAN
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - JIAN LIU
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| |
Collapse
|
22
|
Singh AK, Kumar Pathak A, Kumar P, Kumar Singh A, Kumar Sah Gond M, Singh Negi R, Das R, Agrawal S, Kumar Mishra S, Tiwari KN. Effects of Asiatic acid on brain cancer by altering astrocytes and the AKT1-PRKCB signaling pathway: A genomic and network pharmacology perspective. Brain Res 2025; 1859:149652. [PMID: 40252893 DOI: 10.1016/j.brainres.2025.149652] [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: 01/31/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
The most common primary brain tumor, glioblastoma (GBM), currently has a dismal prognosis because of its fast growth and dissemination. Recent research indicates that Asiatic acid (AA), which is extracted from Trema orientalis L., has potential as a medicinal agent. AA, which was obtained from a methanolic extract of Trema orientalis L. and examined utilizing high-resolution mass spectroscopy (HRMS) analysis, was employed in this investigation. Then, in order to forecast the therapeutic advantages of AA in managing GBM, we conducted an in silico study. Online web servers like SwissADME, pKCSM, and Protox-II were used to assess AA. Then, the major targets of the AA (from Swiss Target Prediction and TargetNet) and GBM (from GeneCards and DisGeNET) were identified. The important genes were then merged into the STRING and ShinyGo databases to examine the protein-protein interaction (PPI) network, gene annotation, and KEGG pathways, with the goal of identifying the core mechanisms involved in GBM management. The top five hub gene targets of the built network (AKT1, SRC, IL-6, TNF, and EGFR) were investigated, along with some contemporaneous additional major targets (PRKCB, GSK3B, ITGB1, BRAF, and PTPN6). These targets were tightly linked to GO activities such as synoviocyte proliferation, cytokine activity, and EGFR tyrosine kinase inhibitor resistance, as well as proteoglycans in cancer-related pathways. Furthermore, a survival study was conducted to assess the chronicity of targets, as well as molecular docking activity between important targets and AA against GBM to determine binding effectiveness. Overall, the study found that AKT1 is the most powerful receptor for AA, having a binding energy of -8.19 kcal/mol, followed by PRKCB (-7.53 kcal/mol). Finally, docking studies suggest that AA has the potential to be an effective treatment for GBM. Furthermore, clinical studies will provide more precise insights into the AA's efficacy as a medicine in the future.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Department of Pharmacognosy, Kunwar Haribansh Singh College of Pharmacy, Jaunpur, Uttar Pradesh 222182, India
| | - Adarsh Kumar Pathak
- Department of Pharmaceutical Chemistry, Ashok Singh Pharmacy College, Jaunpur, Uttar Pradesh 222180, India
| | - Pradeep Kumar
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Anand Kumar Singh
- Department of Chemistry, PG College, Mariahu, VBS Purvanchal University, Jaunpur, Uttar Pradesh 222161, India
| | - Manjeet Kumar Sah Gond
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Rohit Singh Negi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Richa Das
- Department of Biotechnology, Parul Institute of Applied Science, Parul University, Vadodara 391760 Gujarat, India
| | - Shreni Agrawal
- Department of Biotechnology, Parul Institute of Applied Science, Parul University, Vadodara 391760 Gujarat, India
| | - Sunil Kumar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| | - Kavindra Nath Tiwari
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
23
|
Chen Q, Zhuang S, Chen S, Wu B, Zhou Q, Wang W. Targeting the dual miRNA/BMP2 network: LncRNA H19-mediated temozolomide resistance unveils novel therapeutic strategies in glioblastoma. Front Oncol 2025; 15:1577221. [PMID: 40297808 PMCID: PMC12034693 DOI: 10.3389/fonc.2025.1577221] [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: 02/15/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Long noncoding RNA (lncRNA) is known to not only be involved in various biological processes but also to play a crucial role in chemotherapy resistance. The development of resistance in glioblastoma (GBM) poses a significant challenge in clinical settings. Nonetheless, the mechanisms through which lncRNA contributes to acquired resistance to Temozolomide (TMZ) in GBM patients remain unclear. Methods We identified 265 upregulated and 396 downregulated lncRNAs associated with chemoresistance in GBM from the GEO database (GSE100736). Subsequently, we assessed the expression levels of lncRNA H19, hsa-miR-138-5p, hsa-miR-22-3p, and BMP2 mRNA through quantitative polymerase chain reaction (qPCR) in GBM cells and TMZ-resistant GBM cells. Cell viability and proliferation were evaluated using CCK-8 and cell colony formation assays, respectively. Apoptosis was determined through flow cytometry analysis. The impact of gene overexpression and knockdown on cell proliferation and apoptosis was examined via cell transfection experiments. Furthermore, we investigated the influence of lncRNA H19 on tumor development using an in vivo xenograft tumor model. Results The upregulation of lncRNA H19 was observed in TMZ-resistant GBM cell lines and tissues, suggesting its involvement in acquired TMZ resistance. Silencing lncRNA H19 restored TMZ sensitivity in resistant GBM cells in vitro. Conversely, overexpression of lncRNA H19 promoted GBM cell proliferation and hindered TMZ-triggered apoptosis, facilitating the acquisition of TMZ resistance. Notably, lncRNA H19 functions as a molecular decoy for hsa-miR-138-5p and hsa-miR-22-3p, and these miRNAs can reverse the acquired TMZ resistance induced by lncRNA H19 in GBM cells. Additionally, BMP2 gene expression is crucial in the lncRNA H19-mediated pathway of acquired TMZ resistance in GBM cells. Knockdown of lncRNA H19 reinstated TMZ sensitivity in vivo, whereas BMP2 overexpression reinstated TMZ resistance. Conclusion LncRNA H19 enhances TMZ resistance in glioblastoma through competitive RNA targeting of BMP2.
Collapse
Affiliation(s)
- Qiudan Chen
- Department of Clinical Laboratory, Central Laboratory, Jing’an District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Shihao Zhuang
- Department of Pediatrics, Fujian Children’s Hospital, Fuzhou, China
| | - Shuying Chen
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Biying Wu
- Department of Clinical Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Qingyu Zhou
- Department of Clinical Laboratory, Central Laboratory, Jing’an District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Weifeng Wang
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
24
|
Li Y, Wang Y, Han X, Xu J, Liu E, Cheng J, Ma Y, Yang T, Wu J, Sun H, Fan K, Shen D, Li J, Chen X, Yu S, Shu H. Glioma-derived SPARCL1 promotes the formation of peritumoral neuron-glioma synapses. J Neurooncol 2025:10.1007/s11060-025-05007-y. [PMID: 40227556 DOI: 10.1007/s11060-025-05007-y] [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: 01/25/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Gliomas are the most common type of primary malignant brain tumor with high degree of malignancy and rapid progression, and patients often have complications such as epilepsy and cognitive impairment. Thus, identifying related therapeutic targets, prolonging patient survival time and improving patient quality of life are urgently needed. Recent studies have shown that glutamatergic neurons around tumors and glioma cells form synapses, neuron-glioma synapses (NGSs), which have electrophysiological properties and participate in the proliferation, infiltration and invasion of tumors. Therefore, we aimed to explore the molecular mechanisms underlying NGS formation. METHODS We used bioinformatic analysis to screen for the expression of SPARCL1, which may play a role in promoting NGS formation, and we evaluated clinical samples through immunofluorescence, Western blot, and reverse transcriptase polymerase chain reaction (RT‒PCR) assays to validate the bioinformatic analysis results. Vitro neuron-glioma cell coculture model was established and allowed us to edit SPARCL1 expression in glioma cells, further allowing us to investigate the role of SPARCL1 in NGS formation. RESULTS Bioinformatic analysis revealed that SPARCL1 is highly expressed in glioma cells and is associated with synaptogenesis. Clinical samples were evaluated to verify the bioinformatics results, and SPARCL1 was found to be highly distributed in the tumor peripheral region. In the vitro neuron-glioma cell coculture model, NGSs were clearly observed, and SPARCL1 overexpression promoted NGS formation. CONCLUSION Taken together, these findings suggest that SPARCL1 is one of the molecules that promotes NGS formation in the tumor peripheral region.
Collapse
Affiliation(s)
- Yang Li
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Yao Wang
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xingyue Han
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jing Xu
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Enyu Liu
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jingmin Cheng
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yuan Ma
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Tao Yang
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jianping Wu
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Haodong Sun
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Kexia Fan
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Danyi Shen
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jie Li
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xin Chen
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Sixun Yu
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Haifeng Shu
- Department of Neurosurgery, The General Hospital of Western Theater Command, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
25
|
Eibl T, Goschütz F, Liebert A, Ritter L, Steiner HH, Schebesch KM, Neher M. Risk factors for unintended discontinuation of tumor-specific treatment after tumor surgery in glioblastoma patients aged 70 or older. BRAIN & SPINE 2025; 5:104253. [PMID: 40276266 PMCID: PMC12020898 DOI: 10.1016/j.bas.2025.104253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/27/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
Purpose The most beneficial treatment option for newly diagnosed glioblastoma is maximum safe resection and adjuvant therapy. Elderly patients carry a higher perioperative risk for complications, thus, predictors of unfavorable surgical outcome must be evaluated more intensively. Consequently, we sought to evaluate surgery-related paradigms leading to discontinuation of adjuvant treatment after initial neurosurgical resection. Methods Patients receiving microsurgical tumor resection for newly diagnosed glioblastoma CNS WHO grade 4 were evaluated. Further inclusion criteria was age >70 years. Comorbidities were summarized using the Charlson Comorbidity Index (CCI), the 5 and 11 item modified frailty index (mFI-5 and mFI-11) and the CHA2DS2-VASc Score. Primary endpoint was discontinuation of tumor-specific before completion of adjuvant radiotherapy or radio-chemotherapy. Results 102 patients were included, mean age was 76.2 ± 4.2 years. The median extent of contrast-enhancing tumor volume was 99.1 ± 5.9 %. Surgical morbidity and mortality prohibited beginning of adjuvant treatment in 19 patients (18.6 %) and overall discontinuation of treatment before completion of radiotherapy was observed in 26/87 patients (29.9 %). Treatment failure was associated with increasing patient age (p = 0.04) and greater comorbidity scores. The mFI-5 and mFI-11 outperformed the CCI and the CHA2DS2-VASc Score. Two or more points in the 5- and 11-item mFI were significantly associated with increased risk of treatment failure (p = 0.004 and p = 0.001, respectively). Conclusion In Glioblastoma patients, advanced age and comorbidities are relevant confounders and put patients at risk for surgery-related morbidity. Nevertheless, it can be aimed at a maximum safe resection with acceptable surgical morbidity.
Collapse
Affiliation(s)
- Thomas Eibl
- Department of Neurosurgery, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Bavaria, Germany
| | - Franziska Goschütz
- Department of Neurosurgery, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Bavaria, Germany
| | - Adrian Liebert
- Department of Neurosurgery, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Bavaria, Germany
| | - Leonard Ritter
- Department of Neurosurgery, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Bavaria, Germany
| | - Hans-Herbert Steiner
- Department of Neurosurgery, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Bavaria, Germany
| | - Karl-Michael Schebesch
- Department of Neurosurgery, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Bavaria, Germany
| | - Markus Neher
- Department of Neurosurgery, Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Bavaria, Germany
| |
Collapse
|
26
|
Li Y, Liang J, Ren X, Guo J, Wang X, Wang X, Yu S, Li T, Yang X. FGFR3-TACC3 fusion gene promotes glioblastoma malignant progression through the activation of STAT3 signaling pathway. Front Oncol 2025; 15:1560008. [PMID: 40265014 PMCID: PMC12011601 DOI: 10.3389/fonc.2025.1560008] [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: 01/13/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025] Open
Abstract
Objective The Fibroblast growth factor receptors 3-transforming acidic coiled-coil-containing protein 3 (FGFR3-TACC3, F3-T3) oncogenic fusion gene, identified in malignant tumors such as gliomas and bladder cancer, has been particularly noted in recurrent gliomas where it is considered to drive malignant progression, thus presenting itself as a viable therapeutic target. However, the precise mechanism by which F3-T3 facilitates the malignant progression of glioma is not fully understood. Methods Correction analysis of STAT3 and FGFR3 with major glioma mutation types and pan-cancer analysis was conducted using The Cancer Genome Atlas (TCGA) database. A series of phenotypic experiments, including CCK-8, EdU, colony-formation assay, wound healing assay, and transwell assay were conducted to detect the effects of F3-T3 on proliferation, invasion, and migration of glioma cells. The association between F3-T3 and epithelial-mesenchymal transition (EMT) was investigated through enrichment analysis of the E-MTAB-6037 gene chip database and confirmed by western blot. The underling mechanism were further inferred and validated through RNA sequencing, E-MTAB-6037 gene chip data, and western blot. The relationship between p-STAT3 expression and the WHO grade of glioma was evaluated using immunohistochemistry (IHC) and tissue microarray analysis. Furthermore, the results of vivo experiments and IHC has confirmed the impact of F3-T3 on glioma malignant progression and activation of the STAT3 signaling pathway. Results The experimental results from this study indicate that F3-T3 accelerates the epithelial-mesenchymal transition (EMT) process in glioma cells, thereby promoting their proliferation, invasion, and migration capabilities. Mechanistically, it was determined through RNA sequencing that the signal transducer and activator of transcription 3 (STAT3) signaling pathway is crucial for the malignant progression of F3-T3. This finding was further supported through follow-up experiments conducted after STAT3 knockdown. The role of the STAT3 pathway in gliomas was also reinforced through bioinformatic analysis and immunohistochemistry (IHC) on tissue microarrays (TMA). Further in vivo experiments corroborated the role of F3-T3 in enhancing glioma growth and progression. Conclusion F3-T3 facilitates the proliferation, invasion, migration and EMT of glioma cells, thereby promoting their malignant progression through STAT3 signaling activation. These findings highlight its potential as a therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Yiming Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianshen Liang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Xiude Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiahe Guo
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Xisen Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Xuya Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
27
|
Bellavita R, Barra T, Braccia S, Prisco M, Valiante S, Lombardi A, Leone L, Pisano J, Esposito R, Nastri F, D’Errico G, Falanga A, Galdiero S. Engineering Multifunctional Peptide-Decorated Nanofibers for Targeted Delivery of Temozolomide across the Blood-Brain Barrier. Mol Pharm 2025; 22:1920-1938. [PMID: 40091203 PMCID: PMC11979881 DOI: 10.1021/acs.molpharmaceut.4c01125] [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: 10/03/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
A nanoplatform based on self-assembling peptides was developed with the ability to effectively transport and deliver a wide range of moieties across the blood-brain barrier (BBB) for the treatment of glioblastoma. Its surface was functionalized to have a targeted release of TMZ thanks to the targeting peptide that binds to EGFRvIII, which is overexpressed on tumor cells, and gH625, which acts as an enhancer of penetration. Furthermore, the on-demand release of TMZ was achieved through matrix metalloproteinase-9 (MMP-9) cleavage. Nanofibers were characterized for their stability, critical aggregation concentration, and morphology. Next, the effect on both 2D and 3D glioblastoma/astrocytoma (U-87) and glioma (U-118) cell lines was evaluated. The Annexin V/Propidium iodide showed an increase in necrotic and apoptotic cells, and the morphological analysis allowed to discover that both U-118 and U-87 spheroids are smaller in surface, perimeter, and Feret's diameter when treated with NF-TMZ. The developed nanofiber was demonstrated to permeate the BBB in vitro in a 3D spheroidal biodynamic BBB model. Finally, there were no cytotoxic effects of nanofibers without the drug on spheroids, while a significant decrease in viability was observed when NF-TMZ was used. Overall, these results open new opportunities for the evaluation of the efficacy and safety of this nanoplatform in in vivo studies.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department
of Pharmacy, School of Medicine, University
of Naples Federico II, Napoli 80131, Italy
| | - Teresa Barra
- Department
of Biology, University of Napoli Federico
II, Via Cintia, Naples 80126, Italy
| | - Simone Braccia
- Department
of Pharmacy, School of Medicine, University
of Naples Federico II, Napoli 80131, Italy
| | - Marina Prisco
- Department
of Biology, University of Napoli Federico
II, Via Cintia, Naples 80126, Italy
| | - Salvatore Valiante
- Department
of Biology, University of Napoli Federico
II, Via Cintia, Naples 80126, Italy
| | - Assunta Lombardi
- Department
of Biology, University of Napoli Federico
II, Via Cintia, Naples 80126, Italy
| | - Linda Leone
- Department
of Chemical Sciences, University of Napoli
Federico II and 4CSGI (Unit of Naples), Via Cintia, Naples 80126, Italy
| | - Jessica Pisano
- Department
of Biology, University of Napoli Federico
II, Via Cintia, Naples 80126, Italy
| | - Rodolfo Esposito
- Department
of Chemical Sciences, University of Napoli
Federico II and 4CSGI (Unit of Naples), Via Cintia, Naples 80126, Italy
| | - Flavia Nastri
- Department
of Chemical Sciences, University of Napoli
Federico II and 4CSGI (Unit of Naples), Via Cintia, Naples 80126, Italy
| | - Gerardino D’Errico
- Department
of Chemical Sciences, University of Napoli
Federico II and 4CSGI (Unit of Naples), Via Cintia, Naples 80126, Italy
- CSGI
(Unit of Naples), Via
Cintia, Naples 80126, Italy
| | - Annarita Falanga
- Department
of Agricultural Science, University of Naples
Federico II, Via Università
100, Portici, Portici 80055, Italy
| | - Stefania Galdiero
- Department
of Pharmacy, School of Medicine, University
of Naples Federico II, Napoli 80131, Italy
| |
Collapse
|
28
|
Wang L, Xu P, Li X, Zhang Q. Comprehensive bioinformatics analysis identified HMGB3 as a promising immunotherapy target for glioblastoma multiforme. Discov Oncol 2025; 16:478. [PMID: 40192954 PMCID: PMC11977083 DOI: 10.1007/s12672-025-02235-6] [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: 12/16/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE Glioblastoma multiforme (GBM) presents significant therapeutic challenges due to its heterogeneous tumorigenicity, drug resistance, and immunosuppression. Although several molecular markers have been developed, there still lack of sensitive molecular for accurately detection. Studying the mechanisms underlying the development of GBM and finding relevant prognostic biomarkers remains crucial. METHODS Single-cell RNA sequencing, bulk RNA-seq, and cancer immune cycle activities of GBM were used to assess the expression of different molecular related to GBM. Bioinformatics analyses were carried to evaluate the functional of the high mobility group protein B3 (HMGB3) in GBM. RESULTS HMGB3 was highly expressed in GBM tissues and influenced the interpatient and intratumoral transcriptomic heterogeneity as well as immunosuppression in GBM. HMGB3 also contributes to a no inflamed tumor microenvironment (TME) and has an inhibitory effect on tumor-associated immune cell infiltration. Besides, HMGB3 participated GBM chemotherapeutic sensitivity and negative correlation with 140 medicines. CONCLUSION HMGB3 as a heterogeneous and immunosuppressive molecule in the GBM TME, making it a potential target for precision therapy for GBM.
Collapse
Affiliation(s)
- Libin Wang
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
- Medical Research Center, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
| | - Peizhi Xu
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
- Department of Neurosurgery, The 6th Affiliated Hospital of Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
| | - Xinglong Li
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
- Medical Research Center, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
| | - Qinghua Zhang
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
| |
Collapse
|
29
|
Qiu Q, Deng H, Song P, Liu Y, Zhang M. Lactylation in Glioblastoma: A Novel Epigenetic Modifier Bridging Epigenetic Plasticity and Metabolic Reprogramming. Int J Mol Sci 2025; 26:3368. [PMID: 40244246 PMCID: PMC11989911 DOI: 10.3390/ijms26073368] [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: 01/21/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Glioblastoma, the most common and aggressive primary malignant brain tumor, is characterized by a high rate of recurrence, disability, and lethality. Therefore, there is a pressing need to develop more effective prognostic biomarkers and treatment approaches for glioblastoma. Lactylation, an emerging form of protein post-translational modification, has been closely associated with lactate, a metabolite of glycolysis. Since the initial identification of lactylation sites in core histones in 2019, accumulating evidence has shown the critical role that lactylation plays in glioblastoma development, assessment of poor clinical prognosis, and immunosuppression, which provides a fresh angle for investigating the connection between metabolic reprogramming and epigenetic plasticity in glioblastoma cells. The objective of this paper is to present an overview of the metabolic and epigenetic roles of lactylation in the expanding field of glioblastoma research and explore the practical value of developing novel treatment plans combining targeted therapy and immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Mengxian Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.Q.); (H.D.); (P.S.); (Y.L.)
| |
Collapse
|
30
|
Lee SH, Hofstede RP, Noriega de la Colina A, Gunton JH, Bernstock JD, Traverso G. Implantable systems for neurological chronotherapy. Adv Drug Deliv Rev 2025; 221:115574. [PMID: 40187646 DOI: 10.1016/j.addr.2025.115574] [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: 12/01/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Implantable systems for neurological chronotherapy are poised to revolutionize the treatment of central nervous system diseases and disorders. These devices enable precise, time-controlled drug delivery aligned with the body's circadian rhythms, optimizing therapeutic outcomes. By bypassing the blood-brain barrier, they achieve high local drug concentrations while minimizing systemic side effects, offering significant advantages for conditions where traditional therapies often fall short. Platforms like SynchroMed II and CraniUS showcase this innovation, providing programmable delivery for conditions such as epilepsy and glioblastoma, with customizable profiles ranging from continuous infusion to timed bolus administration. Preclinical and clinical studies underscore the efficacy of aligning drug delivery with circadian rhythms, enhancing outcomes in chrono-chemotherapy and anti-epileptic treatments. Despite their promise, challenges remain, including the invasiveness of implantation within the brain, device longevity, synchronization complexities, and cost(s). Accordingly, this review explores the current state of implantable neurological systems that may be leveraged for chronotherapy, their applications, limitations, and potential to transform neurological disease/disorder management.
Collapse
Affiliation(s)
- Seung Ho Lee
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roemer Pott Hofstede
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - John H Gunton
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua D Bernstock
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
31
|
Fan B, Pan Q, Yuan X, Du W, Yan Z. EIF2S2 as a prognostic marker and therapeutic target in glioblastoma: insights into its role and downstream mechanisms. Cancer Cell Int 2025; 25:126. [PMID: 40176031 PMCID: PMC11967041 DOI: 10.1186/s12935-025-03762-6] [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: 10/20/2024] [Accepted: 03/20/2025] [Indexed: 04/04/2025] Open
Abstract
Glioblastoma (GBM) the most common and most aggressive primary brain tumor has a five-year survival rate of less than 5%. The onset of GBM is very complicated and has always been the focus of researchers. This study analyzed data from 637 GBM and 20 normal tissues from The Cancer Genome Atlas (TCGA), and patients were categorized into high and low EIF2S2 expression groups. The Overall survival and disease-free survival of GBM patients in low expression of EIF2S2 group were significantly higher than those in high expression of EIF2S2 group (p < 0.001), and the expression level of EIF2S2 was significantly correlated with tumor grade (p < 0.001) and tumor recurrence (p < 0.001). The study designed three different short hairpin RNA (shRNA) sequence vectors, identifying shEIF2S2-1 as the most effective. This vector significantly reduced EIF2S2 expression, cell proliferation, and migration while increasing apoptosis in SHG-44 and U251 cells (p < 0.01). By detecting SHG-44 cells infected with shEIF2S2 vector and shCtrl with human whole gene expression chip, we identified WNT5A that is a downstream target gene of EIF2S2. Interfering with WNT5A and overexpressing EIF2S2 in SHG-44 and U251 cells revealed that EIF2S2 regulates WNT5A expression. This regulation led to an increased apoptosis rate (p < 0.05) and a significant reduction in cell migration (p < 0.05) in both the EIF2S2 overexpression and shWNT5A interference groups, confirming that WNT5A is a downstream regulatory target of EIF2S2. This study revealed the key role of EIF2S2 in GBM and its potential molecular mechanism.
Collapse
Affiliation(s)
- Bo Fan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Qing Pan
- Department of Hemodialysis, The Second Hospital of Hebei Medical University, No.215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Xiaokai Yuan
- Department of Rehabilitation Medicine, The Second Hospital of Hebei Medical University, No.215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Wei Du
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Zhongjie Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China.
| |
Collapse
|
32
|
Jiang X, Lai Y, Xia W, Yang W, Wang J, Pan J, Zhao Q, Zhou F, Li S, Zhang S, Gao J, Wang Y, Zan T, Xu ZP, Yu H, Xu Z. Self-Oxygenating PROTAC Microneedle for Spatiotemporally-Confined Protein Degradation and Enhanced Glioblastoma Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411869. [PMID: 40025927 DOI: 10.1002/adma.202411869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/31/2024] [Indexed: 03/04/2025]
Abstract
Glioblastoma (GBM) is the most aggressive subtype of primary brain tumors, which marginally respond to standard chemotherapy due to the blood-brain barrier (BBB) and the low tumor specificity of the therapeutics. Herein, a double-layered microneedle (MN) patch is rationally engineered by integrating acid and light dual-activatable PROteolysis TArgeting Chimera (PROTAC) nanoparticles and self-oxygenating BSA-MnO2 (BM) nanoparticles for GBM treatment. The MN is administrated at the tumor site to locally deliver the PROTAC prodrug and BM nanoparticles. The PROTAC nanoparticles are rapidly released from the outer layer of the MN and specifically activated in the acidic intracellular environment of tumor cells. Subsequently, near-infrared light activates the photosensitizer to produce singlet oxygen (1O2) through photodynamic therapy (PDT), thereby triggering spatiotemporally-tunable degradation of bromodomain and extraterminal protein 4 (BRD4). The BM nanoparticles, in the inner layer of the MN, serve as an oxygen supply station, and counteracts tumor hypoxia by converting hydrogen peroxide (H2O2) into oxygen (O2), thus promoting PDT and PROTAC activation. This PROTAC prodrug-integrated MN significantly inhibits tumor growth in both subcutaneous and orthotopic GBM tumor models. This study describes the first spatiotemporally-tunable protein degradation strategy for highly efficient GBM therapy, potentially advancing precise therapy of other kinds of refractory brain tumors.
Collapse
Affiliation(s)
- Xingyu Jiang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yi Lai
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenzheng Xia
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wenfang Yang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Junjue Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiaxing Pan
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qian Zhao
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Feng Zhou
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shiqin Li
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shunan Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Jing Gao
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhi Ping Xu
- Institute of Biomedical Health Technology and Engineering and Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518067, China
| | - Haijun Yu
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
33
|
Grabenweger R, Völz D, Bumes E, Best M, Paal P. 'Nurses as Gatekeepers': Nurses' Responses to Spiritual Needs of Patients with Primary Malignant Brain Tumors in Austria-Analysis of a Qualitative Vignette Study. JOURNAL OF RELIGION AND HEALTH 2025; 64:732-753. [PMID: 39984792 PMCID: PMC11950041 DOI: 10.1007/s10943-025-02278-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 02/23/2025]
Abstract
This paper investigates the nurses' gatekeeping role for spiritual care in multi-disciplinary teams as a way of responding to the spiritual needs of patients with brain tumors in Austria. Using a validated vignette in an online survey, qualitative data from 56 neurosurgical nurses (30.4% response rate) were analyzed via reflexive thematic analysis. One key theme, Nurses as Gatekeepers - Referral and Working with other Health Care Professionals, highlights nurses' critical role in spiritual care. Effective referrals depend on nurses' awareness of spiritual care specialists' roles, emphasizing their importance in ensuring comprehensive, team-based care for neuro-oncological patients with complex spiritual needs.
Collapse
Affiliation(s)
- Reinhard Grabenweger
- Institute of Nursing Science and Practice, Paracelsus Medical University, Strubergasse 21, Salzburg, 5020, Austria.
| | - Daniela Völz
- Department of Neurology and Wilhelm Sander - NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Elisabeth Bumes
- Department of Neurology and Wilhelm Sander - NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Megan Best
- Institute for Ethics and Society, University of Notre Dame Australia, Broadway, Australia
| | - Piret Paal
- Department of Ethnology, Institute of Cultural Studies, Tartu University, Tartu, Estonia
- Institute of Palliative Care, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
34
|
Chang X, Guo H, Li Y, Ding J. Implantable devices for resected glioblastoma therapy. Asian J Pharm Sci 2025; 20:101034. [PMID: 40182136 PMCID: PMC11964529 DOI: 10.1016/j.ajps.2025.101034] [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: 06/13/2024] [Revised: 11/30/2024] [Accepted: 12/26/2024] [Indexed: 04/05/2025] Open
Abstract
Glioblastoma (GBM) is a highly infiltrative brain tumor. The treatment of GBM is challenging due to the existence of blood brain barrier, its highly invasive nature, and its heterogeneity. Given the limitations of conventional therapies, this Perspective explores the development trajectory of implantable devices, highlighting the advantages of current models. With the progression in research, these implantable devices certainly hold promising potential for GBM therapy.
Collapse
Affiliation(s)
- Xiaoyu Chang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun 130061, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Hui Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yunqian Li
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun 130061, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
35
|
Li Y, Ta L, Wu Q, Zhang H, Xu Y, Gan L, Liu J. Assessment of the Validity of Carbon Ion Irradiation for C6 Gliomas in Rats. Dose Response 2025; 23:15593258251327505. [PMID: 40297665 PMCID: PMC12033543 DOI: 10.1177/15593258251327505] [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: 07/15/2024] [Revised: 01/15/2025] [Accepted: 02/18/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose Application of energy-spectrum computed tomography (CT) to assess specific efficacy of and response to carbon ion radiotherapy (CIRT) of C6 gliomas in rats. Methods After establishing C6 glioma rat models, 3 tumor-bearing rats were randomly selected as controls. The remaining were divided into 0 Gy, 1 Gy, and 2 Gy groups for CIRT. Energy-spectrum CT scans were performed, and brain tissues were collected for histopathology and western blot Test. Survival rates in each group were compared. Results The results demonstrated that tumors in the 1 Gy and 2 Gy groups decreased at different rates up to 14 days post-CIRT (P < 0.05). Furthermore, compared to pre-CIRT measurements, the energy-spectrum parameters gradually increased in the 0 Gy group, while they decreased in the 2 Gy group. Post-CIRT, the Ki-67 proliferation index and the expression levels of vascu-larassociated proteins in tumor tissues were significantly reduced in the 1 and 2 Gy groups. Additionally, the survival times of tumor-bearing rats were prolonged after CIRT. Conclusions CIRT effectively restricts tumor cell growth and proliferation, leading to improved survival rates in rats with C6 gliomas. The use of energy-spectrum CT with immunohistochemistry for quantitative detection can actively support the effectiveness of carbon ion radiotherapy in inhibiting tumor proliferation.
Collapse
Affiliation(s)
- Yufeng Li
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Lei Ta
- Ningbo Medical Center LiHuiLi Hospital, Ningbo, China
| | - QingFeng Wu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Hongyu Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yuan Xu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Lu Gan
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Jianli Liu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
36
|
Wu Y, Chen Z, Shi M, Qiu S, Zhang Y. Nimotuzumab and bevacizumab combined with temozolomide and radiotherapy in patients with newly diagnosed glioblastoma multiforme: a retrospective single-arm study. J Neurooncol 2025; 172:429-436. [PMID: 39760795 DOI: 10.1007/s11060-024-04932-8] [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: 11/17/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE Glioblastoma (GBM), the most common malignant tumor of the central nervous system (CNS) in adults, continues to result in poor survival rates despite standard treatment. Advancements in understanding GBM's molecular complexity have increased interest in targeted therapeutic approaches. This retrospective, single-center, single-arm study combined nimotuzumab and bevacizumab with radiotherapy (RT) and temozolomide (TMZ) for the treatment of newly diagnosed GBM. The objectives were to determine the efficacy of this treatment combination and the associated toxicity. METHODS A retrospective analysis of clinical data of GBM patients treated at our institution from September 2021 to May 2023 with postoperative combination therapy of nimotuzumab, bevacizumab, and TMZ concurrent with RT, as well as maintenance therapy with bevacizumab and TMZ. Follow-ups were performed every 3 to 6 months via hospital visits and telephone interviews. The primary endpoints were overall survival (OS) and progression-free survival (PFS). The secondary endpoint was the incidence of adverse events (AEs). RESULTS A total of 18 patients were included. The median follow-up time was 23 months. The one-year PFS rate was 77.8%, and the one-year OS rate was 94.4%. The median PFS was 18 months (95%CI, 15.9-20.1), and the median OS was 28 months (95%CI, 18.9-37.1). All AEs were controllable. CONCLUSION The combination of nimotuzumab and bevacizumab with TMZ and RT appears to demonstrate efficacy and safety in newly diagnosed GBM patients, providing a reference for clinical treatment. Further prospective studies are needed to confirm our results.
Collapse
Affiliation(s)
- Yaping Wu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Zhiying Chen
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Mingtao Shi
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Shuo Qiu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Yongchun Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China.
| |
Collapse
|
37
|
Yang C, Ma C, Xu C, Li S, Li C, Wang Z, Li Z. Comprehensive Evaluation of Frailty and Sarcopenia Markers to Predict Survival in Glioblastoma Patients. J Cachexia Sarcopenia Muscle 2025; 16:e13809. [PMID: 40234099 PMCID: PMC11999731 DOI: 10.1002/jcsm.13809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common primary malignant brain tumour in adults. Patients with GBM are particularly susceptible to moderate-to-high frail. Frailty status has been associated with the outcome of many types of cancer, including GBM, although there is still little consensus regarding the specific criteria for assessing frailty status. This study aimed to determine the predictive significance of the modified frailty score (mFS) in GBM patients using haematological and sarcopenia indicators. METHODS Between January 2016 and September 2022, we enrolled 309 adult GBM patients. Data on demographics, haematological examination, and temporal muscle thickness (TMT) were collected and assessed. The prognostic relevance of the frailty parameters was established using Kaplan-Meier and Cox proportional model. The scoring systems were created by integrating these indicators. Variables with independent prognostic values were used to construct the nomograms. Nomogram accuracy was evaluated using the calibration curve, Harrell's concordance index (C-index), and time-dependent receiver operating characteristic curves. Clinical practicality was assessed using decision curve analysis. RESULTS The baseline characteristics of the 309 participants revealed a median age of 59 years (interquartile range 52-66) with a predominance of male patients (58.58%). TMT (hazard ratio [HR] = 3.787, 95% confidence interval [CI] 2.576-5.566, p < 0.001), prognostic nutritional index (HR = 1.722, 95% CI 1.098-2.703, p = 0.018), and mean corpuscular volume (HR = 1.958, 95% CI 1.111-3.451, p = 0.020) were identified as independent prognostic markers. The constructed mFS, obtained by integrating these three indices, exhibited independent prognostic significance (HR = 2.461, 95% CI 1.751-3.457, p < 0.001). The patients in the low-risk group had a median overall survival (OS) of 13.9 months, while the patients in the high risk had a median OS of 5.8 months. Importantly, the mFS demonstrated significant independent prognostic value in the subgroup aged > 65 (HR = 1.822, 95% CI 1.011-3.284, p = 0.046). The nomogram, which included the mFS, demonstrated high accuracy, with a c-index of 0.781. The nomogram bootstrapped calibration plot also performed well compared to the ideal model. Nomograms showed promising discriminative potential, with time-dependent areas under the curves of 0.945, 0.835, and 0.820 for 0.5-, 1-, and 2-year overall survival prediction, respectively. CONCLUSIONS Preoperative mFS is a comprehensive frailty marker for predicting survival outcomes in patients with GBM. A dynamic nomogram incorporating the mFS may facilitate preoperative survival evaluation. Early and appropriate multimodal interventions, including nutritional support, rehabilitation, and psychological care, may help in the neurosurgical care of patients with GBM or other brain tumours.
Collapse
Affiliation(s)
- Chao Yang
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Chao Ma
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Cheng‐Shi Xu
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Si‐Rui Li
- Department of RadiologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Chen Li
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Ze‐Fen Wang
- Department of PhysiologyWuhan University School of Basic Medical SciencesWuhanChina
| | - Zhi‐Qiang Li
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Clinical NutritionZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
38
|
Chang CW, Bale A, Bhargava R, Harley BA. Glioblastoma drives protease-independent extracellular matrix invasion of microglia. Mater Today Bio 2025; 31:101475. [PMID: 39896278 PMCID: PMC11787038 DOI: 10.1016/j.mtbio.2025.101475] [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: 10/04/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025] Open
Abstract
Glioblastoma (GBM) is the most common and lethal form of primary brain cancer. Microglia infiltration into the tumor microenvironment is associated with immunosuppression and poor prognosis. Improved physicochemical understanding of microglia activation and invasion may provide novel GBM therapeutic strategies essential for improving long-term treatment efficacy. Here, we combine microfluidic systems with 3-D collagen hydrogels to systematically investigate microglia activation, invasion, contractility and cytokine secretion in response to GBM-microglia crosstalk. GBM inflammatory biomolecules significantly promote activation and 3D invasion of microglia. Interestingly, microglia invasion is not significantly affected by inhibitors of MMP activity or cellular glycolysis. In contrast, ROCK-pathway inhibition significantly impedes microglia invasion. Infrared microscopy analyses show that GBM conditioned media does not significantly alter microglia lipid content. Further, GBM conditioned media resulted in significantly increased collagen hydrogel contraction, suggesting the importance of microglia contractility to physically remodel the local extracellular matrix (ECM). We also identify a panel of soluble proteins that may contribute to microglia chemotaxis, such as TIMP-1 and CXCL12. Taken together, this study suggests that the presence of GBM cells can enhance microglia invasion via increased cellular contractility, independent of MMP activity and cellular glycolysis.
Collapse
Affiliation(s)
- Chia-Wen Chang
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Ashwin Bale
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Rohit Bhargava
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- CZ Biohub Chicago, LLC, Chicago, IL 60642, USA
| | - Brendan A.C. Harley
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
39
|
Ramachandran R, Jeans AF. Breaking Down Glioma-Microenvironment Crosstalk. Neuroscientist 2025; 31:177-194. [PMID: 39066464 PMCID: PMC11909767 DOI: 10.1177/10738584241259773] [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: 07/28/2024]
Abstract
High-grade gliomas (HGGs) are the commonest primary brain cancers. They are characterized by a pattern of aggressive growth and diffuse infiltration of the host brain that severely limits the efficacy of conventional treatments and patient outcomes, which remain generally poor. Recent work has described a suite of mechanisms via which HGGs interact, predominantly bidirectionally, with various cell types in the host brain including neurons, glial cells, immune cells, and vascular elements to drive tumor growth and invasion. These insights have the potential to inspire novel approaches to HGG therapy that are critically needed. This review explores HGG-host brain interactions and considers whether and how they might be exploited for therapeutic gain.
Collapse
|
40
|
Gao Y, Zhang M, Wang G, Lai W, Liao S, Chen Y, Ning Q, Tang S. Metabolic cross-talk between glioblastoma and glioblastoma-associated microglia/macrophages: From basic insights to therapeutic strategies. Crit Rev Oncol Hematol 2025; 208:104649. [PMID: 39922398 DOI: 10.1016/j.critrevonc.2025.104649] [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: 01/06/2025] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Glioblastoma (GBM), a highly malignant "cold" tumor of the central nervous system, is characterized by its ability to remodel the GBM immune microenvironment (GME), leading to significant resistance to immunotherapy. GBM-associated microglia/macrophages (GAMs) are essential components of the GME. Targeting GAMs has emerged as a promising strategy against GBM. However, their highly immunosuppressive nature contributes to GBM progression and drug resistance, significantly impeding anti-GBM immunotherapy. Accumulating evidence suggests that metabolic reprogramming accompanies GBM progression and GAM polarization, which are in turn driven by specific metabolic abnormalities and altered cellular signaling pathways. Importantly, metabolic crosstalk between GBM and GAMs further promotes tumor progression. Clarifying and disrupting this metabolic crosstalk is expected to enhance the antitumor phenotype of GAMs and inhibit GBM malignant progression. This review explores metabolism-based interregulation between GBM and GAMs and summarizes recent therapeutic strategies targeting this crosstalk, offering new insights into GBM immunotherapy.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Mengxia Zhang
- Department of Histology and Embryology, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Guihua Wang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Weiwei Lai
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Shuxian Liao
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Yao Chen
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| | - Shengsong Tang
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
41
|
Angerstein AO, Young LEA, Thanasupawat T, Vriend J, Grimsley G, Lun X, Senger DL, Sinha N, Beiko J, Pitz M, Hombach-Klonisch S, Drake RR, Klonisch T. Distinct spatial N-glycan profiles reveal glioblastoma-specific signatures. J Pathol 2025; 265:486-501. [PMID: 39967571 DOI: 10.1002/path.6401] [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: 07/29/2024] [Revised: 11/21/2024] [Accepted: 12/24/2024] [Indexed: 02/20/2025]
Abstract
This study explored the complex interactions between glycosylation patterns, tumour biology, and therapeutic responses to temozolomide (TMZ) in human malignant glioma, specifically CNS WHO grade 3 oligodendroglioma (ODG) and glioblastoma (GB). Using spatial imaging of N-glycans in formalin-fixed paraffin-embedded (FFPE) tissue sections via MALDI-MSI, we analysed the N-glycome in primary and recurrent GB tissues and orthotopic xenografts of patient-derived brain tumour-initiating cells (BTIC) sensitive or resistant to TMZ. We identified unique N-glycosylation profiles, with nontumor brain (NTB) and ODG showing higher levels of bisecting and tri-antennary structures, while GB exhibited more tetra-antennary and sialylated N-glycans. Distinctive sialylation patterns were observed, with specific α2,6 and α2,3 isomeric linkages significantly altered in GB. Moreover, comparative analysis of primary and recurrent GB tissues revealed elevated high mannose N-glycans in primary GB and fucosylated bi- and tri-antennary N-glycans in recurrent GB tissues. Next, in the orthotopic xenografts of TMZ-sensitive and TMZ-resistant patient brain tumour initiating cells (BTIC), we identified potential N-glycan markers for TMZ treatment response and resistance. Finally, we found significantly altered expression of genes involved in N-glycan biosynthesis in malignant glioma, highlighting the crucial role of N-glycans in glioma and therapy resistance. This study lays the foundation for developing glycosylation-based diagnostic biomarkers and targeted therapies, potentially improving clinical outcomes for GB patients. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Aaron O Angerstein
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Lyndsay E A Young
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Thatchawan Thanasupawat
- Department of Human Anatomy and Cell Science, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
| | - Jerry Vriend
- Department of Human Anatomy and Cell Science, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
| | - Grace Grimsley
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Xueqing Lun
- Cumming School of Medicine, Arnie Charbonneau Cancer Institute, Calgary, AB, Canada
| | - Donna L Senger
- Cumming School of Medicine, Arnie Charbonneau Cancer Institute, Calgary, AB, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Montreal, QC, Canada
| | - Namita Sinha
- Department of Pathology, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
| | - Jason Beiko
- Department of Surgery, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
| | - Marshall Pitz
- Department of Internal Medicine, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
- Department of Pathology, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
- Department of Pathology, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
42
|
Zhou J. Construction of enhanced MRI-based radiomics models using machine learning algorithms for non-invasive prediction of IL7R expression in high-grade gliomas and its prognostic value in clinical practice. J Transl Med 2025; 23:383. [PMID: 40165301 PMCID: PMC11959755 DOI: 10.1186/s12967-025-06402-9] [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: 12/28/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND High-grade gliomas are among the most aggressive and deadly brain tumors, highlighting the critical need for improved prognostic markers and predictive models. Recent studies have identified the expression of IL7R as a significant risk factor that affects the prognosis of patients diagnosed with high-grade gliomas (HGG). This research focuses on investigating the prognostic significance of Interleukin 7 Receptor (IL7R) expression and aims to develop a noninvasive predictive model based on radiomics for HGG. METHODS We conducted an analysis using data from The Cancer Genome Atlas (TCGA) and The Cancer Imaging Archive (TCIA), focusing on a group of 310 patients diagnosed with high-grade gliomas. To evaluate prognosis, we applied both univariate and multivariate Cox regression analyses alongside Kaplan-Meier survival analysis. Radiomics features were extracted from specific regions of interest, which were outlined by two physicians using 3D Slicer software. For selecting the most relevant features, we utilized the Minimum Redundancy Maximum Relevance (mRMR) and Recursive Feature Elimination (RFE) algorithms. Following this, we developed and assessed Support Vector Machine (SVM) and Logistic Regression (LR) models, measuring their performance through various metrics such as accuracy, specificity, sensitivity, positive predictive value, calibration curves, the Hosmer-Lemeshow goodness-of-fit test, decision curve analysis (DCA), and Kaplan-Meier survival analysis. RESULTS The survival analysis encompassed a total of 310 patients diagnosed with high-grade glioma, sourced from the TCGA database. Patients were stratified into high and low expression groups based on the levels of IL7R expression. Kaplan-Meier survival curves and Cox regression analysis revealed that an increase in IL7R expression correlated with a decline in overall survival (OS). The median Intraclass Correlation Coefficient (ICC) for the assessed radiomic features was determined to be 0.869, with 93 features exhibiting an ICC of 0.75 or greater. Utilizing the mRMR and RFE methodologies led to the identification of a final set comprising eight features. The Support Vector Machine (SVM) model recorded an Area Under the Curve (AUC) value of 0.805, whereas the AUC derived from fivefold cross-validation was noted to be 0.768. Conversely, the Logistic Regression (LR) model produced an AUC of 0.85, with an internal fivefold cross-validation AUC of 0.779, indicating a more robust predictive capability. We developed Support Vector Machine (SVM) and Logistic Regression (LR) models, with the LR model demonstrating a more robust predictive capability. Further Kaplan-Meier analysis underscored a significant association between elevated risk scores from the LR model and OS malignancy, with a P value of less than 0.001. GSVA analysis showed the enrichment pathway of KEGG and Hallmark genes in the high RS group. Moreover, expression levels of the LOX gene and the infiltration of M0 macrophages were significantly heightened in the high-risk score group, alongside an increase in tumor mutation burden (TMB). Interestingly, the mutation frequencies of TP53 and PIK3CA were found to be lower in the high-risk score group when compared to their low-risk counterparts. CONCLUSION IL7R expression is a vital prognostic marker in high-grade gliomas. The radiomics-based LR models demonstrate strong predictive capabilities for patient outcomes. Future investigations should aim to incorporate these insights into clinical practice to enhance personalized treatment approaches for patients with high-grade glioma.
Collapse
Affiliation(s)
- Jie Zhou
- Shenzhen Hospital (Longgang), Beijing University of Chinese Medicine, Shenzhen, China.
| |
Collapse
|
43
|
Wang Y, Hu Z, Wang H. The clinical implications and interpretability of computational medical imaging (radiomics) in brain tumors. Insights Imaging 2025; 16:77. [PMID: 40159380 PMCID: PMC11955438 DOI: 10.1186/s13244-025-01950-6] [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: 10/17/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Radiomics has widespread applications in the field of brain tumor research. However, radiomic analyses often function as a 'black box' due to their use of complex algorithms, which hinders the translation of brain tumor radiomics into clinical applications. In this review, we will elaborate extensively on the application of radiomics in brain tumors. Additionally, we will address the interpretability of handcrafted-feature radiomics and deep learning-based radiomics by integrating biological domain knowledge of brain tumors with interpretability methods. Furthermore, we will discuss the current challenges and prospects concerning the interpretability of brain tumor radiomics. Enhancing the interpretability of radiomics may make it more understandable for physicians, ultimately facilitating its translation into clinical practice. CRITICAL RELEVANCE STATEMENT: The interpretability of brain tumor radiomics empowers neuro-oncologists to make well-informed decisions from radiomic models. KEY POINTS: Radiomics makes a significant impact on the management of brain tumors in several key clinical areas. Transparent models, habitat analysis, and feature attribute explanations can enhance the interpretability of traditional handcrafted-feature radiomics in brain tumors. Various interpretability methods have been applied to explain deep learning-based models; however, there is a lack of biological mechanisms underlying these models.
Collapse
Affiliation(s)
- Yixin Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Zongtao Hu
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Hongzhi Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China.
| |
Collapse
|
44
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [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: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
45
|
Fu M, Xue B, Miao X, Gao Z. Overcoming immunotherapy resistance in glioblastoma: challenges and emerging strategies. Front Pharmacol 2025; 16:1584688. [PMID: 40223940 PMCID: PMC11987931 DOI: 10.3389/fphar.2025.1584688] [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: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults, characterized by rapid proliferation, extensive infiltration, and significant intratumoral heterogeneity. Despite advancements in conventional treatments, including surgery, radiotherapy, and chemotherapy, the prognosis for GBM patients remains poor, with a median survival of approximately 15 months. Immunotherapy has emerged as a promising alternative; however, the unique biological and immunological features, including its immunosuppressive tumor microenvironment (TME) and low mutational burden, render it resistant to many immunotherapeutic strategies. This review explores the key challenges in GBM immunotherapy, focusing on immune evasion mechanisms, the blood-brain barrier (BBB), and the TME. Immune checkpoint inhibitors and CAR-T cells have shown promise in preclinical models but have limited clinical success due to antigen heterogeneity, immune cell exhaustion, and impaired trafficking across the BBB. Emerging strategies, including dual-targeting CAR-T cells, engineered immune cells secreting therapeutic molecules, and advanced delivery systems to overcome the BBB, show potential for enhancing treatment efficacy. Addressing these challenges is crucial for improving GBM immunotherapy outcomes.
Collapse
Affiliation(s)
- Maowu Fu
- Department of Neurosurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Bing Xue
- Department of Neurosurgery, Jinan Third People’s Hospital, Jinan, Shandong, China
| | - Xiuming Miao
- Department of Pathology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zong Gao
- Department of Neurosurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
46
|
Senatore E, Avolio R, Rinaldi L, Chiuso F, Oliva MA, D'Ambrosio C, Bianco AG, Dalla E, Pagnotta SM, Flammia R, Ambrosino C, Memoli D, Turacchio G, Mimoune SI, Toiron Y, Audebert S, Camoin L, Lignitto L, Scaloni A, Arcella A, Feliciello A. Praja2 controls P-body assembly and translation in glioblastoma by non-proteolytic ubiquitylation of DDX6. EMBO Rep 2025:10.1038/s44319-025-00425-5. [PMID: 40148504 DOI: 10.1038/s44319-025-00425-5] [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: 10/08/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal form of malignant brain tumor in adults. Dysregulation of protein synthesis contributes to cancer cell plasticity, driving GBM cell heterogeneity, metastatic behavior, and drug resistance. Understanding the complex network and signaling pathways governing protein translation, is therefore an important goal for GBM treatment. Here we identify a novel signaling network centered on the E3 ubiquitin ligase praja2 that controls protein translation in GBM. Praja2 forms a multimeric complex with the RNA helicase DDX6, which inhibits translation of target RNAs within processing bodies (P-bodies). Stimulation of cAMP signaling through activation of G-protein-coupled receptors induces P-body assembly through praja2-mediated non-proteolytic polyubiquitylation of DDX6. Genetic inactivation of praja2 reshapes DDX6/mRNA complexes and translating polysomes and promotes cellular senescence and GBM growth arrest. Expression of an ubiquitylation-defective DDX6 mutant suppresses the assembly of P-bodies and sustains GBM growth. Taken together, our findings identify a cAMP-driven network that controls translation in P-bodies and GBM growth.
Collapse
Affiliation(s)
- Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Rosario Avolio
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | - Chiara D'Ambrosio
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici, Naples, Italy
| | - Antonio Giuseppe Bianco
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Emiliano Dalla
- Department of Medicine, University of Udine, Udine, Italy
| | | | - Raffaella Flammia
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Concetta Ambrosino
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Domenico Memoli
- Department of Medicine, Surgery and Dentistry SMS, University of Salerno, Salerno, Italy
| | - Gabriele Turacchio
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Sonia Ines Mimoune
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Yves Toiron
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Stephane Audebert
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Luc Camoin
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Luca Lignitto
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici, Naples, Italy
| | | | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy.
| |
Collapse
|
47
|
Tang Q, Ma C, Xie J, Zhang Q, Zhang B, Bian W, Lu Q, Wan Z, Wu W. Unraveling anoikis in glioblastoma: insights from single-cell sequencing and prognostic modeling. Cancer Cell Int 2025; 25:116. [PMID: 40140848 PMCID: PMC11948803 DOI: 10.1186/s12935-025-03752-8] [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: 07/20/2024] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Despite advances, Glioblastoma (GBM) treatment remains challenging due to its rapid progression and resistance to therapies. OBJECTIVES This study aimed to investigate the role of anoikis-a mechanism by which cells evade programmed cell death upon detachment from the extracellular matrix-in GBM progression and prognosis. METHODS Utilizing single-cell sequencing and bulk-transcriptome sequencing data from TCGA, GEO, and CGGA databases, we performed comprehensive bioinformatics analyses. We identified anoikis-related genes, constructed a prognostic model using 101 machine learning algorithms, and validated its clinical utility across multiple cohorts.Finally, we also verified the expression of model genes and the function of key gene in clinical samples and cell lines. RESULTS Single-cell sequencing revealed heterogeneous expression of anoikis-related genes across distinct cell populations within GBM. MES-like Malignant cells and Myeloids exhibited higher enrichment of these genes, implicating their role in anoikis resistance and tumor aggressiveness. The prognostic model, based on identified genes, effectively stratified patients into high-risk and low-risk groups, demonstrating significant differences in survival outcomes. Mutation and tumor microenvironment analyses highlighted distinct genetic landscapes and immune cell infiltration patterns associated with different risk groups. SLC43A3 emerged as a key gene, showing significant upregulation in tumor tissues and correlating with poor prognosis in GBM. CONCLUSION This study provides insights into the molecular mechanisms of anoikis resistance in GBM, underscoring its critical role in tumor progression and patient prognosis. The developed prognostic model offers a promising tool for personalized treatment strategies and warrants further exploration of targeted therapies to improve outcomes for GBM patients.
Collapse
Affiliation(s)
- Qikai Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Chenfeng Ma
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Qixiang Zhang
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Bingtao Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, People's Republic of China
| | - Weiqi Bian
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Qingyu Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Zeyu Wan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
48
|
Shen H, Zhou Z, Zhang X, Xu M, Jiang X, Qin W, Chen S. Comparative analysis of pyrosequencing and next-generation sequencing for assessing MGMT methylation in glioma patients. J Neurooncol 2025:10.1007/s11060-025-05015-y. [PMID: 40138094 DOI: 10.1007/s11060-025-05015-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND The methylation status of the O6-methylguanine-DNA methyltransferase (MGMT) gene promoter is pivotal in clinical decision-making for glioma patients. Pyrosequencing (PSQ) has been regarded as the gold standard for determining the MGMT promoter status. Nevertheless, PSQ is limited by its low throughput, high costs, and intricate protocols. In this study, we present a comparative analysis of the performance of PSQ and next-generation sequencing (NGS) in evaluating MGMT methylation in glioma patients. METHODS Initially, we developed an amplicon-based NGS method for quantifying MGMT methylation. Subsequently, a comparative assessment was carried out to evaluate the MGMT promoter methylation levels in 50 formalin-fixed paraffin-embedded (FFPE) glioma samples using both PSQ and NGS. Finally, a consistency analysis was performed to compare the results obtained from PSQ and NGS. RESULTS The results revealed a significant correlation between PSQ and NGS (R2 = 0.88). Moreover, the consistency rate of the test results among the 50 samples was 94% (47/50), with one negative sample and two positive samples showing inconsistency. These three samples were verified using MethyLight technology, and the results were consistent with those obtained from NGS. CONCLUSIONS This study indicates that, although PSQ is the gold standard, the quantitative detection of MGMT methylation by NGS is more accurate than that by PSQ. NGS is characterized by high throughput and cost-effectiveness, while also yielding accurate and stable results. Therefore, NGS provides a viable alternative to the PSQ method for detecting MGMT methylation.
Collapse
Affiliation(s)
- Huanming Shen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- HaploX Biotechnology, Shenzhen, China
| | | | | | | | | | - Wenjian Qin
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Shifu Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- HaploX Biotechnology, Shenzhen, China.
| |
Collapse
|
49
|
Stea DM, D’Alessio A. Caveolae: Metabolic Platforms at the Crossroads of Health and Disease. Int J Mol Sci 2025; 26:2918. [PMID: 40243482 PMCID: PMC11988808 DOI: 10.3390/ijms26072918] [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: 02/27/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Caveolae are small flask-shaped invaginations of the plasma membrane enriched in cholesterol and sphingolipids. They play a critical role in various cellular processes, including signal transduction, endocytosis, and mechanotransduction. Caveolin proteins, specifically Cav-1, Cav-2, and Cav-3, in addition to their role as structural components of caveolae, have been found to regulate the activity of signaling molecules. A growing body of research has highlighted the pivotal role of caveolae and caveolins in maintaining cellular metabolic homeostasis. Indeed, studies have demonstrated that caveolins interact with the key components of insulin signaling, glucose uptake, and lipid metabolism, thereby influencing energy production and storage. The dysfunction of caveolae or the altered expression of caveolins has been associated with metabolic disorders, including obesity, type 2 diabetes, and ocular diseases. Remarkably, mutations in caveolin genes can disrupt cellular energy balance, promote oxidative stress, and exacerbate metabolic dysregulation. This review examines current research on the molecular mechanisms through which caveolae and caveolins regulate cellular metabolism, explores their involvement in the pathogenesis of metabolic disorders, and discusses potential therapeutic strategies targeting caveolin function and the stabilization of caveolae to restore metabolic homeostasis.
Collapse
Affiliation(s)
- Dante Maria Stea
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Alessio D’Alessio
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Rome, Italy
| |
Collapse
|
50
|
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] [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
|