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
|
Saeed MU, Choudhury A, Ansari J, Mohammad T, Hussain A, Fatima U, Alajmi MF, Hassan MI. Discovering potential therapeutic targets in glioblastoma multiforme using a multi-omics approach. Pathol Res Pract 2025; 269:155942. [PMID: 40168776 DOI: 10.1016/j.prp.2025.155942] [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: 10/27/2024] [Revised: 03/08/2025] [Accepted: 03/09/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor associated with high fatality rates, poor prognosis, and limited treatment options. In this study, we utilized RNA-Seq gene count data from GBM patients, sourced from the Gene Expression Omnibus (GEO) database, to conduct an in-depth analysis of gene expression patterns. METHODS Our investigation involved stratifying samples into two distinct sets, Group I and Group II, comparing normal, low-grade, and GBM tumor samples, respectively. Subsequently, we performed differential expression analysis and enrichment analysis to uncover significant gene signatures. To elucidate the protein-protein interactions associated with GBM, we used the STRING plugin within Cytoscape for comprehensive network visualization and analysis. RESULTS By applying Maximal clique centrality (MCC) scores, we identified a set of 10 hub genes in each group. These hub genes were subjected to survival analysis, highlighting their prognostic relevance. In Group I, comprising BUB1, DLGAP5, BUB1B, CDK1, TOP2A, CDC20, KIF20A, ASPM, BIRC5, and CCNB2, these genes emerged as potential biomarkers associated with the transition to low-grade tumors. In Group II, genes such as LIF, LBP, CSF3, IL6, CCL2, SAA1, CCL20, MMP9, CXCL10, and MMP1 were found to be involved in the transformation to adult glioblastoma. Kaplan-Meier's overall survival analysis of these hub genes revealed that modifications, particularly the upregulation of these candidate genes, were associated with reduced survival in GBM patients. CONCLUSIONS The findings established the significance of genomic alterations and differential gene expression in GBM, presenting opportunities for prognostic and targeted therapeutic interventions. This study provides valuable insights into potential avenues for enhancing the clinical management of GBM.
Collapse
Affiliation(s)
- Mohammad Umar Saeed
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Jaoud Ansari
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Urooj Fatima
- UAMS, Donald W. Reynolds Institute on Aging, Department of Geriatrics, Little Rock, AR 72205, USA
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
3
|
Chithra P, Bhatia D, Solanki R. Advanced nanomicelles for targeted glioblastoma multiforme therapy. BIOMATERIALS ADVANCES 2025; 170:214221. [PMID: 39922136 DOI: 10.1016/j.bioadv.2025.214221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and malignant primary brain tumor, classified as grade IV by the WHO. Despite standard treatments like surgical resection, radiotherapy and chemotherapy (i.e. temozolomide), GBM's prognosis remains poor due to its heterogeneity, recurrence and the impermeability of the blood-brain barrier (BBB). The exact cause of GBM is unclear with potential factors including genetic predisposition and ionizing radiation. Innovative approaches such as nanomicelles-nanoscale, self-assembled structures made from lipids and amphiphilic polymers show promise for GBM therapy. These nanocarriers enhance drug solubility and stability, enabling targeted delivery of therapeutic agents across the BBB. This review explores the synthesis strategies, characterization and applications of nanomicelles in GBM treatment. Nanomicelles improve the delivery of both hydrophobic and hydrophilic drugs and provide non-invasive delivery options. By offering site-specific targeting, biocompatibility, and stability, nanomicelles can potentially overcome the limitations of current GBM therapies. This review highlights recent advancements in the use of nanomicelles for delivering therapeutic agents and nucleic acids addressing the critical need for advanced treatments to improve GBM patient outcomes.
Collapse
Affiliation(s)
- P Chithra
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
4
|
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
|
5
|
Zhang X, Ou N, Liu C, Zhuo Z, Matthews PM, Liu Y, Ye C, Bai W. Unsupervised brain MRI tumour segmentation via two-stage image synthesis. Med Image Anal 2025; 102:103568. [PMID: 40199108 DOI: 10.1016/j.media.2025.103568] [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: 09/13/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Deep learning shows promise in automated brain tumour segmentation, but it depends on costly expert annotations. Recent advances in unsupervised learning offer an alternative by using synthetic data for training. However, the discrepancy between real and synthetic data limits the accuracy of the unsupervised approaches. In this paper, we propose an approach for unsupervised brain tumour segmentation on magnetic resonance (MR) images via a two-stage image synthesis strategy. This approach accounts for the domain gap between real and synthetic data and aims to generate realistic synthetic data for model training. In the first stage, we train a junior segmentation model using synthetic brain tumour images generated by hand-crafted tumour shape and intensity models, and employs a validation set with distribution shift for model selection. The trained junior model is applied to segment unlabelled real tumour images, generating pseudo labels that capture realistic tumour shape, intensity, and texture. In the second stage, realistic synthetic tumour images are generated by mixing brain images with tumour pseudo labels, closing the domain gap between real and synthetic images. The generated synthetic data is then used to train a senior model for final segmentation. In experiments on five brain imaging datasets, the proposed approach, named as SynthTumour, surpasses existing unsupervised methods and demonstrates high performance for both brain tumour segmentation and ischemic stroke lesion segmentation tasks.
Collapse
Affiliation(s)
- Xinru Zhang
- School of Integrated Circuits and Electronics, Beijing Institute of Technology, Beijing, China; Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Ni Ou
- School of Automation, Beijing Institute of Technology, Beijing, China
| | - Chenghao Liu
- School of Integrated Circuits and Electronics, Beijing Institute of Technology, Beijing, China
| | - Zhizheng Zhuo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, United Kingdom; UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Chuyang Ye
- School of Integrated Circuits and Electronics, Beijing Institute of Technology, Beijing, China.
| | - Wenjia Bai
- Department of Brain Sciences, Imperial College London, London, United Kingdom; Department of Computing, Imperial College London, London, United Kingdom.
| |
Collapse
|
6
|
Xue X, He Z, Liu F, Wang Q, Chen Z, Lin L, Chen D, Yuan Y, Huang Z, Wang Y. Taurochenodeoxycholic acid suppresses the progression of glioblastoma via HMGCS1/HMGCR/GPX4 signaling pathway in vitro and in vivo. Cancer Cell Int 2025; 25:160. [PMID: 40264142 PMCID: PMC12016240 DOI: 10.1186/s12935-025-03782-2] [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: 10/24/2024] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the foremost prevalent and highly aggressive intracranial malignancy, which urgently needs safer and more efficacious therapeutic strategies. Our research aimed to investigate the impact and the underlying mechanism of Taurochenodeoxycholic acid (TCDCA) on GBM. In this study, we explored the suppressive effect of TCDCA in vitro by qualification of proliferation and migration assays and flow cytometry, and subsequently predicted the potential anti-GBM mechanism of TCDCA by mRNA sequencing and the following rescue experiments. An orthotopic GBM model in C57BL/6 mice further demonstrated the anti-GBM mechanism of TCDCA. In vitro experiments verified that TCDCA inhibited the growth and migration of GBM cells and induced cell cycle arrest at the G2/M phase. Subsequent mechanism investigations showed that upregulation of HMGCS1 and HMGCR and downregulation of glutathione peroxidase-4 (GPX4) was observed in GBM cells by TCDCA treatment. Notably, inhibitory effects of proliferation and migration as well as induction of ferroptosis by TCDCA were partially restored by Simvastatin (SIN), a competitive HMGCR inhibitor. Furthermore, TCDCA showed an anti-GBM effect in an orthotopic transplantation model in vivo. TCDCA impedes GBM progression by virtue of this intricately orchestrated molecular cascade, through HMGCS1/HMGCR/GPX4 signaling axis, thus unveiling a novel therapeutic avenue warranting further scrutiny in the treatment landscape of GBM.
Collapse
Affiliation(s)
- Xiumin Xue
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Ziwan He
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Furui Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Qian Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhichao Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Lin Lin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Danni Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yinfeng Yuan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
7
|
Liu X, Abmanhal-Masarweh H, Iwanowytsch O, Okwelogu E, Arashvand K, Karabatsou K, Ivo D'Urso P, Roncaroli F, Kostarelos K, Kisby T, Hadjidemetriou M. Plasma-to-tumour tissue integrated proteomics using nano-omics for biomarker discovery in glioblastoma. Nat Commun 2025; 16:3412. [PMID: 40210624 PMCID: PMC11986092 DOI: 10.1038/s41467-025-58252-0] [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: 10/14/2024] [Accepted: 03/17/2025] [Indexed: 04/12/2025] Open
Abstract
Glioblastoma (GB) is the most lethal brain cancer, with patient survival rates remaining largely unchanged over the past two decades. Here, we introduce the Nano-omics integrative workflow that links systemic (plasma) and localised (tumour tissue) protein changes associated with GB progression. Mass spectrometry analysis of the nanoparticle biomolecule corona in GL261-bearing mice at different stages of GB revealed plasma protein alterations, even at low tumour burden, with over 30% overlap between GB-specific plasma and tumour tissue proteomes. Analysis of matched plasma and surgically resected tumour samples from high-grade glioma patients demonstrates the clinical applicability of the Nano-omics pipeline. Cross-species correlation identified 48 potential GB biomarker candidates involved in actin cytoskeleton organisation, focal adhesion, platelet activation, leukocyte migration, amino acid biosynthesis, carbon metabolism, and phagosome pathways. The Nano-omics approach holds promise for the discovery of early detection and disease monitoring biomarkers of central nervous system conditions, paving the way for subsequent clinical validation.
Collapse
Affiliation(s)
- Xinming Liu
- NanoOmics Lab, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NanoTherapeutics Lab, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Hanan Abmanhal-Masarweh
- NanoOmics Lab, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Olivia Iwanowytsch
- NanoOmics Lab, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Emmanuel Okwelogu
- NanoOmics Lab, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Kiana Arashvand
- NanoTherapeutics Lab, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Konstantina Karabatsou
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester, UK
| | - Pietro Ivo D'Urso
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester, UK
| | - Federico Roncaroli
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, UK
| | - Kostas Kostarelos
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, UK
- Nanomedicine Lab, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Barcelona, Spain
- Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys, Barcelona, Spain
| | - Thomas Kisby
- NanoTherapeutics Lab, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, UK.
| | - Marilena Hadjidemetriou
- NanoOmics Lab, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Centre for Nanotechnology in Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, UK.
| |
Collapse
|
8
|
Liu H, Hu W, Zhang L, Li Z, Liu J, Chen L. Plasma-Derived Exosomal i-tRF-LeuCAA as Biomarker for Glioma Diagnosis and Promoter of Epithelial-Mesenchymal Transition via TPM4 Regulation. CNS Neurosci Ther 2025; 31:e70356. [PMID: 40202170 PMCID: PMC11979793 DOI: 10.1111/cns.70356] [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: 11/18/2024] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
AIMS This study aimed to discover plasma-derived exosomal tsRNAs that serve as novel diagnostic biomarkers for glioma and to investigate the mechanism by which tsRNAs regulate glioma development. METHODS Differentially expressed tsRNAs in the plasma exosomes of glioma patients were identified using small RNA array sequencing. Bioinformatics analyses were used to predict the biological function of tsRNAs. The changes in the phenotypes of glioma cells treated with a tsRNA mimic and inhibitor were detected. The diagnostic and prognostic characteristics of potential target genes and their related functions in gliomas were further analyzed. The cell and animal experiments were used to analyze the molecular mechanisms. RESULTS Among the 453 differentially expressed tsRNAs identified in the plasma-derived exosomes of glioma patients using small RNA sequencing, i-tRF-LeuCAA was associated with the prognosis and molecular diagnostic characteristics of glioma patients and promoted the migration, invasion, and proliferation of glioma cells and inhibited their apoptosis. In addition, TPM4 is a potential target of i-tRF-LeuCAA and is related to epithelial-mesenchymal transition in gliomas. CONCLUSIONS i-tRF-LeuCAA could be served as a non-invasive biomarker in the diagnosis and prognosis of glioma. i-tRF-LeuCAA may indirectly regulate TPM4 expression and influence epithelial-mesenchymal transition, which may promote glioma progression.
Collapse
Affiliation(s)
- Hongyu Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryHainan Hospital of Chinese PLA General HospitalSanyaHainanChina
- Department of NeurosurgeryFirst Medical Center of Chinese PLA General HospitalBeijingChina
| | - Wentao Hu
- School of Medicine, Nankai UniversityTianjinChina
| | - Lijun Zhang
- Laboratory of OncologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Ze Li
- Department of NeurosurgeryFirst Medical Center of Chinese PLA General HospitalBeijingChina
| | - Jialin Liu
- Department of NeurosurgeryFirst Medical Center of Chinese PLA General HospitalBeijingChina
| | - Ling Chen
- Department of NeurosurgeryFirst Medical Center of Chinese PLA General HospitalBeijingChina
| |
Collapse
|
9
|
Shamul JG, Wang Z, Gong H, Ou W, White AM, Moniz-Garcia DP, Gu S, Clyne AM, Quiñones-Hinojosa A, He X. Meta-analysis of the make-up and properties of in vitro models of the healthy and diseased blood-brain barrier. Nat Biomed Eng 2025; 9:566-598. [PMID: 39304761 PMCID: PMC11922799 DOI: 10.1038/s41551-024-01250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
In vitro models of the human blood-brain barrier (BBB) are increasingly used to develop therapeutics that can cross the BBB for treating diseases of the central nervous system. Here we report a meta-analysis of the make-up and properties of transwell and microfluidic models of the healthy BBB and of BBBs in glioblastoma, Alzheimer's disease, Parkinson's disease and inflammatory diseases. We found that the type of model, the culture method (static or dynamic), the cell types and cell ratios, and the biomaterials employed as extracellular matrix are all crucial to recapitulate the low permeability and high expression of tight-junction proteins of the BBB, and to obtain high trans-endothelial electrical resistance. Specifically, for models of the healthy BBB, the inclusion of endothelial cells and pericytes as well as physiological shear stresses (~10-20 dyne cm-2) are necessary, and when astrocytes are added, astrocytes or pericytes should outnumber endothelial cells. We expect this meta-analysis to facilitate the design of increasingly physiological models of the BBB.
Collapse
Affiliation(s)
- James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Zhiyuan Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Hyeyeon Gong
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | | | - Shuo Gu
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA
| | | | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA.
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
10
|
Meryem N, Soufiane B, Sanaa EM, Asmae N, Nabil I. Recurrent glioblastoma: a typical example of a complex approach. Rep Pract Oncol Radiother 2025; 30:126-128. [PMID: 40242411 PMCID: PMC11999010 DOI: 10.5603/rpor.104738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/30/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Naciri Meryem
- Department of Radiation Oncology, Cheikh Khalifa University Hospital, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Bouchbouk Soufiane
- Department of Medical Oncology, Cheikh Khalifa University Hospital, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - El Majjaoui Sanaa
- Department of Radiation Oncology, Cheikh Khalifa University Hospital, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Naim Asmae
- Department of Radiation Oncology, Cheikh Khalifa University Hospital, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Ismaili Nabil
- Department of Medical Oncology, Cheikh Khalifa University Hospital, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| |
Collapse
|
11
|
Furuta R, Miyake A. Fibroblast growth factor 22. Differentiation 2025; 143:100860. [PMID: 40139106 DOI: 10.1016/j.diff.2025.100860] [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: 12/23/2024] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Fibroblast growth factor 22 (FGF22) is a member of the FGF7 subfamily that functions as a paracrine factor and was identified in the human placenta in 2001. The FGF22 gene is located on human chromosome 19p13.3, mouse chromosome 10, and zebrafish chromosome 22 and is closely linked to the BSG, HCN2, and POLRMT genes. The gene is composed of three exons, which are common in humans, mice, and zebrafish. However, in humans and mice, FGF22 is produced as two isoforms by alternative splicing, whereas no isoforms have been reported in zebrafish. In humans, FGF22 is expressed in the skin, brain, and ovaries, whereas in mice, it is expressed in the skin, brain, retina, spinal cord, and cochlea. Various abnormalities have been reported in these regions in Fgf22 mutant mice. In zebrafish, fgf22 is expressed in the forebrain, midbrain, and otic vesicles during embryogenesis, and an analysis of knockdown zebrafish models revealed an important role for fgf22 in the process of brain formation. As expected from the results of these functional analyses, FGF22 is also associated with human diseases such as depression, spinal cord injury, hearing loss, and cancer.
Collapse
Affiliation(s)
- Rise Furuta
- Department of Molecular Biology, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1, Shichibancho, Wakayama 640-8156, Japan
| | - Ayumi Miyake
- Department of Molecular Biology, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1, Shichibancho, Wakayama 640-8156, Japan.
| |
Collapse
|
12
|
Latulippe J, Roy LO, Gobeil F, Fortin D. Optimization of Intra-Arterial Administration of Chemotherapeutic Agents for Glioblastoma in the F98-Fischer Glioma-Bearing Rat Model. Biomolecules 2025; 15:421. [PMID: 40149957 PMCID: PMC11940523 DOI: 10.3390/biom15030421] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma (GBM) is a difficult disease to treat for different reasons, with the blood-brain barrier (BBB) preventing therapeutic drugs from reaching the tumor being one major hurdle. The median overall survival is only 14.6 months after the standard first line of treatment. At relapse, there is no recognized standard second-line treatment. Our team uses intra-arterial (IA) chemotherapy as a means to bypass the BBB, hence achieving an overall median survival of 25 months. However, most patients eventually fail the treatment and progress. This is why we wish to expand our portfolio of options in terms of chemotherapy agents available for IA administration. In this study, we tested topotecan, cytarabine, and new formulations of carboplatin and paclitaxel by IA administration in the F98-Fischer glioma-bearing rat model as a screening tool for identifying potential candidate drugs. The topotecan IA group showed increased survival compared to the intravenous (IV) group (29.0 vs. 23.5), whereas the IV cytarabine group survived longer than the IA group (26.5 vs. 22.5). The new formulation of carboplatin showed a significant increase in survival compared to two previous studies with the conventional form (37.5 vs. 26.0 and 30.0). As for paclitaxel, it was too neurotoxic for IA administration. Topotecan and the new formulation of carboplatin demonstrated significant results, warranting their transition for consideration in clinical trials.
Collapse
Affiliation(s)
- Juliette Latulippe
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (J.L.); (F.G.)
| | - Laurent-Olivier Roy
- Division of Neurosurgery, Department of Surgery, Centre Hospitalier de l’Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Fernand Gobeil
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (J.L.); (F.G.)
| | - David Fortin
- Division of Neurosurgery, Department of Surgery, Centre Hospitalier de l’Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| |
Collapse
|
13
|
Wang R, Zhang Y, Guo Y, Zeng W, Li J, Wu J, Li N, Zhu A, Li J, Di L, Cao P. Plant-derived nanovesicles: Promising therapeutics and drug delivery nanoplatforms for brain disorders. FUNDAMENTAL RESEARCH 2025; 5:830-850. [PMID: 40242551 PMCID: PMC11997602 DOI: 10.1016/j.fmre.2023.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/17/2023] [Accepted: 09/10/2023] [Indexed: 04/18/2025] Open
Abstract
Plant-derived nanovesicles (PDNVs), including plant extracellular vesicles (EVs) and plant exosome-like nanovesicles (ELNs), are natural nano-sized membranous vesicles containing bioactive molecules. PDNVs consist of a bilayer of lipids that can effectively encapsulate hydrophilic and lipophilic drugs, improving drug stability and solubility as well as providing increased bioavailability, reduced systemic toxicity, and enhanced target accumulation. Bioengineering strategies can also be exploited to modify the PDNVs to achieve precise targeting, controlled drug release, and massive production. Meanwhile, they are capable of crossing the blood-brain barrier (BBB) to transport the cargo to the lesion sites without harboring human pathogens, making them excellent therapeutic agents and drug delivery nanoplatform candidates for brain diseases. Herein, this article provides an initial exposition on the fundamental characteristics of PDNVs, including biogenesis, uptake process, isolation, purification, characterization methods, and source. Additionally, it sheds light on the investigation of PDNVs' utilization in brain diseases while also presenting novel perspectives on the obstacles and clinical advancements associated with PDNVs.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingjie Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yumiao Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Wei Zeng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jinge Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jie Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Anran Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Peng Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
14
|
Gai S, Yan Q, Li S, Zhong X, Qin Y, Jiang M. Lactoferrin Nanoparticle-Vanadium Complex: A Promising High-Efficiency Agent against Glioblastoma by Triggering Autophagy and Ferroptosis. J Med Chem 2025; 68:4650-4662. [PMID: 39945608 DOI: 10.1021/acs.jmedchem.4c02696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Glioblastoma represents the most aggressive type of brain cancer with minimal clinical advancements in recent decades attributed to the absence of efficient drug delivery strategies. In this study, we synthesized a series of vanadium complexes (V1-V4) and then constructed a lactoferrin (LF)-V4 nanoparticle (NP) delivery system. The nanoplatform crossed the blood-brain barrier by binding to low-density lipoprotein receptor-associated protein-1 and selectively targeted glioblastoma, ultimately inhibiting the growth of in situ glioblastoma tumors. LF-V4 NPs induced autophagic cell death in U87-MG cells by generating reactive oxygen species (ROS) that damaged the mitochondria. Further studies revealed that LF-V4 NPs triggered lipid peroxidation through the accumulation of ROS, the depletion of GSH, and the downregulation of GPX4 and SLC7A11, ultimately leading to ferroptosis in glioblastoma cells.
Collapse
Affiliation(s)
- Shuangshuang Gai
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
- Institute for History and Culture of Science and Technology, Guangxi Minzu University, Nanning 530006, China
| | - Qiwei Yan
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Shan Li
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Xuwei Zhong
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Yiming Qin
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Ming Jiang
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| |
Collapse
|
15
|
Que T, Li Z, Zheng H, Tan JE, Yuan X, Yi G, Fang L, Nie J, Yin Y, Xu H, Zheng X, Liu J, Zhang XA, Qi S, Huang G. Classification of unilateral thalamic gliomas predicts tumor resection and patient's survival: a single center retrospective study. J Neurosurg Sci 2025; 69:111-122. [PMID: 35416455 DOI: 10.23736/s0390-5616.22.05660-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The aim of this study was to propose our classification about unilateral thalamic gliomas, and to describe relationship between the classification and clinical characteristics including symptoms, surgical approaches and survival, which should contribute to the treatment and the prognostic prediction of unilateral thalamic gliomas. METHODS A total of 66 adult unilateral thalamic glioma patients with pathologic confirmation between January 2010 and December 2018 were retrospectively investigated. RESULTS Unilateral thalamic gliomas could be divided into quadrigeminal cistern and ventricle extension type (type Q), lateral type (type L) and anterior type (type A) according to tumor location, extensive polarity and location of ipsilateral posterior limb of internal capsule. Each subtype of QLA classification could match with one kind of corresponding approach. Preoperative symptoms including headache, dyskinesia, aphasia, hydrocephalus and KPS scores, and pathological features including H3K27M mutation and P53 expression were correlated with QLA classification. Further analysis confirmed that type Q tumors had a higher rate of total resection and a significantly longer survival time compared to type L and type A tumors, with similar improved and deteriorated rates of symptoms. Univariate and multivariate analysis demonstrated QLA classification was remarkedly associated with overall survival and could be considered as an independent prognostic factor in patients with unilateral thalamic gliomas. CONCLUSIONS Unilateral thalamic glioma could be divided into 3 subtypes by imaging characteristics, symptoms and survival. QLA classification could predict tumor resection and the prognosis and could contribute to the planning of therapeutic strategy in patients with unilateral thalamic gliomas.
Collapse
Affiliation(s)
- Tianshi Que
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haojie Zheng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Er Tan
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Yuan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Luxiong Fang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Nie
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanyi Yin
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyan Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxuan Zheng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junlu Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xi-An Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China -
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
da Silva KC, Lima IS, dos Santos CC, Nonaka CKV, Souza BSDF, David JM, Ulrich H, do Nascimento RP, Costa MDFD, dos Santos BL, Costa SL. Agathisflavone Inhibits Viability and Modulates the Expression of miR-125b, miR-155, IL-6, and Arginase in Glioblastoma Cells and Microglia/Macrophage Activation. Molecules 2025; 30:158. [PMID: 39795214 PMCID: PMC11721753 DOI: 10.3390/molecules30010158] [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: 11/07/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Glioblastomas (GBM) are malignant tumours with poor prognosis. Treatment involves chemotherapy and/or radiotherapy; however, there is currently no standard treatment for recurrence, and prognosis remains unfavourable. Inflammatory mediators and microRNAs (miRNAs) influence the aggressiveness of GBM, being involved in the communication with the cells of the tumour parenchyma, including microglia/macrophages, and maintaining an immunosuppressive microenvironment. Hence, the modulation of miRNAs and inflammatory factors may improve GBM treatments. In this study, we investigated the effects of agathisflavone, a biflavonoid purified from Cenostigma pyramidale (Tul.), on the growth and migration of GBM cells, on the expression of inflammatory cytokines and microRNAs, as well on the response of microglia. Agathisflavone (5-30 μM) induced a dose- and time-dependent reduction in the viability of both human GL-15 and rat C6 cells, as determined by the MTT test, and reduced cell migration, as determined by cell scratch assay. RT-qPCR analysis revealed that agathisflavone (5 μM) down-regulated the expression of miR-125b and miR-155 in the secretome derived from GL-15 cells, which was associated with upregulation of the mRNA expression of IL-6 and arginase-1 immunoregulatory factors. Exposure of human microglia/macrophage to the secretome from GL-15 GMB cells modulated proliferation and morphology, effects that were modulated by agathisflavone treatment. These results demonstrate the effect of flavonoids on the growth of GBM cells, which impacts cells in the microenvironment and can be considered for preclinical studies for adjuvant treatments.
Collapse
Affiliation(s)
- Karina Costa da Silva
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Irlã Santos Lima
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Carolina Kymie Vasques Nonaka
- Center of Biotechnology and Cell Therapy, São Rafael Hospital, D’Or Institute for Research and Teaching, Salvador 41253-190, BA, Brazil; (C.K.V.N.); (B.S.d.F.S.)
| | - Bruno Solano de Freitas Souza
- Center of Biotechnology and Cell Therapy, São Rafael Hospital, D’Or Institute for Research and Teaching, Salvador 41253-190, BA, Brazil; (C.K.V.N.); (B.S.d.F.S.)
- Institute Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, BA, Brazil
| | - Jorge Mauricio David
- Department of General and Inorganic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40231-300, BA, Brazil;
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748-Butantã, São Paulo 05508-900, SP, Brazil;
| | - Ravena Pereira do Nascimento
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-971, RJ, Brazil
| | - Balbino Lino dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
- College of Nursing, Federal University of Vale do São Francisco, Av. José de Sá Maniçoba, S/N, Petrolina 56304-917, PE, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-971, RJ, Brazil
| |
Collapse
|
17
|
Li J, Liu L, Zong G, Yang Z, Zhang D, Zhao B. Knockdown of CENPF induces cell cycle arrest and inhibits epithelial‑mesenchymal transition progression in glioma. Oncol Lett 2025; 29:61. [PMID: 39611064 PMCID: PMC11602827 DOI: 10.3892/ol.2024.14807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/02/2024] [Indexed: 11/30/2024] Open
Abstract
Gliomas are among the most common malignant tumors of the central nervous system. Despite surgical resection followed by postoperative radiotherapy and chemotherapy, their prognosis remains unfavorable. The present study aimed to assess new mechanisms and explore promising prognostic biomarkers for patients with glioma using comprehensive bioinformatics analysis and in vitro and in vivo assays. Overlapping differentially expressed genes were screened from The Cancer Genome Atlas, GSE111260 and GSE16011 samples for protein-protein interaction networks, a risk score model, gene mutation analysis and a nomogram to identify the prognostic hub genes. Subsequently, an immunoassay was performed to determine key genes. Functional and animal assays were then performed to assess the tumorigenesis of the key genes in glioma. Using bioinformatics analysis, centromere protein F (CENPF), kinesin superfamily member 20A, kinesin superfamily protein 4A and marker of proliferation Ki-67 were identified as potential prognostic biomarkers for patients with glioma. Furthermore, CENPF knockdown was demonstrated to suppress the proliferation and metastasis of glioma cells, and induce G2 arrest in the cell cycle. Moreover, CENPF knockdown was revealed to decrease Vimentin and increase E-cadherin levels in glioma cells, and significantly reduce the size and mass of tumors in vivo. Overall, the present study identified new clinical biomarkers and revealed that CENPF may promote glioma progression by regulating the epithelial-mesenchymal transition pathway. By elucidating the complexities of glioma and identifying prognostic biomarkers, the present research enables further improvement of patient outcomes and the advancement of precision medicine for this disease.
Collapse
Affiliation(s)
- Jia Li
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Lei Liu
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Gang Zong
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Deran Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
18
|
Wu Y, Liu L, Li Z, Zhang T, Wang Q, Cheng M. A Risk Model Based on Ferroptosis-Related Genes OSMR, G0S2, IGFBP6, IGHG2, and FMOD Predicts Prognosis in Glioblastoma Multiforme. CNS Neurosci Ther 2025; 31:e70161. [PMID: 39815665 PMCID: PMC11735466 DOI: 10.1111/cns.70161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/23/2024] [Accepted: 11/24/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a common and highly aggressive brain tumor with a poor prognosis. However, the prognostic value of ferroptosis-related genes (FRGs) and their classification remains insufficiently studied. OBJECTIVE This study aims to explore the significance of ferroptosis classification and its risk model in GBM using multi-omics approaches and to evaluate its potential in prognostic assessment. METHODS Ferroptosis-related genes (FRGs) were retrieved from databases such as FerrDB. The TCGA-GBM and CGGA-GBM datasets were used as training and testing cohorts, respectively. Univariate Cox regression and LASSO regression analyses were performed to establish a risk model comprising five genes (OSMR, G0S2, IGFBP6, IGHG2, FMOD). A Meta-analysis of integrated TCGA and GTEx data was conducted to examine the differential expression of these genes between GBM and normal tissues. Key gene protein expression differences were analyzed using CPTAC and HPA databases. Single-cell RNA sequencing (scRNA-seq) analysis was employed to explore the cell type-specific distribution of these genes. RESULTS The five-gene risk model demonstrated significant prognostic value in GBM. Meta-analysis revealed distinct expression patterns of the identified genes between GBM and normal tissues. Protein expression analysis confirmed these differences. scRNA-seq analysis highlighted the diverse distribution of these genes across different cell types, offering insights into their biological roles. CONCLUSION The ferroptosis-based risk model provides valuable prognostic insights into GBM and highlights potential therapeutic targets, emphasizing the biological significance of ferroptosis-related genes in tumor progression.
Collapse
Affiliation(s)
- Yaqiu Wu
- Department of Neurosurgery Intensive Care Unit, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Ling Liu
- Department of Neurosurgery, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Zhili Li
- Department of Neurosurgery, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Tian Zhang
- Department of Neurosurgery, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Qi Wang
- Department of Neurosurgery, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Meixiong Cheng
- Department of Neurosurgery, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
19
|
Dhiman A, Rana D, Benival D, Garkhal K. Comprehensive insights into glioblastoma multiforme: drug delivery challenges and multimodal treatment strategies. Ther Deliv 2025; 16:87-115. [PMID: 39445563 PMCID: PMC11703381 DOI: 10.1080/20415990.2024.2415281] [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: 05/06/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and malignant brain tumors, with a high prevalence in elderly population. Most chemotherapeutic agents fail to reach the tumor site due to various challenges. However, smart nanocarriers have demonstrated excellent drug-loading capabilities, enabling them to cross the blood brain tumor barrier for the GBM treatment. Surface modification of nanocarriers has significantly enhanced their potential for targeting therapeutics. Moreover, recent innovations in drug therapies, such as the incorporation of theranostic agents in nanocarriers and antibody-drug conjugates, have offered newer insights for both diagnosis and treatment. This review focuses on recent advances in new therapeutic interventions for GBM, with an emphasis on the nanotheranostics systems to maximize therapeutic and diagnostic outcomes.
Collapse
Affiliation(s)
- Ashish Dhiman
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kalpna Garkhal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| |
Collapse
|
20
|
Wang W, Li J, He Q, Liu C, Zheng Z, Zhang B, Mou S, Sun W, Zhao J. Crosstalk between CD180-overexpression macrophages and glioma cells worsens patient survival through malignant phenotype promotion and immunosuppressive regulation. Mol Med 2024; 30:264. [PMID: 39707188 DOI: 10.1186/s10020-024-01029-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Understanding the molecular mechanisms in immunosuppressive regulation is crucial for improving immunotherapeutic strategies. Macrophages, the major immune cells in tumor microenvironment (TME), play a dual role in tumor progression. CD180, primarily expressed in macrophages, remains unclear and requires further investigation. METHODS RNA-seq data were obtained to analyze CD180 expression in gliomas and assess its prognostic value. The comprehensive immune infiltration analysis was performed. Single-cell RNA-seq (scRNA-seq) data were used to examine CD180 expression distribution at the cellular level. CD180-overexpression macrophages were co-cultured with three glioma cell lines. The effects on glioma cell behavior were evaluated through qRT-PCR, Western blot, CCK-8 assay, EdU assay, Transwell assay, TUNEL assay, and flow cytometry. Differentially expressed genes (DEGs) and their potential biological functions were analyzed between different CD180 expression groups. Consensus clustering was used to identify CD180-related glioma subtypes. RESULTS CD180 was significantly upregulated in glioma samples and associated with poor prognosis. High CD180 expression was correlated with increased immune cell infiltration, particularly macrophages, and elevated levels of immune checkpoints. Analysis of scRNA-seq data revealed the predominant expression of CD180 in macrophages within the glioma TME. In vitro experiments demonstrated that CD180-overexpression macrophages promoted glioma cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), while decreasing apoptosis. Mutations in TP53 and PTEN were significantly more prevalent in the high CD180 expression group. We identified nine chemotherapeutic agents that were more effective in glioma patients with high CD180 expression. Additionally, two CD180-related glioma subtypes with distinct prognosis were identified. CONCLUSIONS This study confirmed the prognostic role of CD180 in glioma and its involvement in immunosuppressive regulation and malignant phenotype promotion. Therefore, CD180 was considered as a promising target for immunotherapeutic strategies in glioma treatment.
Collapse
Affiliation(s)
- Wen Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhiyao Zheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Bojian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Siqi Mou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
21
|
Park DH, Bhojnagarwala PS, Liaw K, Bordoloi D, Tursi NJ, Zhao S, Binder ZA, O’Rourke D, Weiner DB. Novel tri-specific T-cell engager targeting IL-13Rα2 and EGFRvIII provides long-term survival in heterogeneous GBM challenge and promotes antitumor cytotoxicity with patient immune cells. J Immunother Cancer 2024; 12:e009604. [PMID: 39622583 PMCID: PMC11624777 DOI: 10.1136/jitc-2024-009604] [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] [Accepted: 09/08/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is known for its high antigenic heterogeneity, which undermines the effectiveness of monospecific immunotherapies. Multivalent immunotherapeutic strategies that target multiple tumor antigens simultaneously could enhance clinical outcomes by preventing antigen-driven tumor escape mechanisms. METHODS We describe novel trivalent antibodies, DNA-encoded tri-specific T-cell engagers (DTriTEs), targeting two GBM antigens, epidermal growth factor receptor variant III (EGFRvIII) and IL-13Rα2, and engaging T cells through CD3. We engineered three DTriTE constructs, each with a unique arrangement of the antigen-binding fragments within a single-chain sequence. We assessed the binding efficiency and cytotoxic activity of these DTriTEs in vitro on target cells expressing relevant antigens. In vivo efficacy was tested in immunocompromised mice, including a longitudinal expression study post-administration and a survival analysis in an NOD scid gamma (NSG)-K mouse model under a heterogeneous tumor burden. RNA sequencing of DTriTE-activated T cells was employed to identify the molecular pathways influenced by the treatment. The antitumor cytotoxicity of patient-derived immune cells was evaluated following stimulation by DTriTE to assess its potential effectiveness in a clinical setting. RESULTS All DTriTE constructs demonstrated strong binding to EGFRvIII and IL-13Rα2-expressing cells, induced significant T cell-mediated cytotoxicity, and enhanced cytokine production (interferon-γ, tumor necrosis factor (TNF)-α, and interleukin(IL)-2). The lead construct, DT2035, sustained expression for over 105 days in vivo and exhibited elimination of tumor burden in a heterogeneous intracranial GBM model, outperforming monospecific antibody controls. In extended survival studies using the NSG-K model, DT2035 achieved a 67% survival rate over 120 days. RNA sequencing of DTriTE-activated T cells showed that DT2035 enhances genes linked to cytotoxicity, proliferation, and immunomodulation, reflecting potent immune activation. Finally, DT2035 effectively induced target-specific cytotoxicity in post-treatment peripheral blood mononuclear cells from patients with GBM, highlighting its potential for clinical effectiveness. CONCLUSIONS DTriTEs exhibit potent anti-tumor effects and durable in vivo activity, offering promising therapeutic potential against GBM. These findings support further development of such multivalent therapeutic strategies to improve treatment outcomes in GBM and potentially other antigenically heterogeneous tumors. The opportunity to advance such important therapies either through biologic delivery or direct in vivo nucleic acid production is compelling.
Collapse
Affiliation(s)
- Daniel H Park
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Kevin Liaw
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Devivasha Bordoloi
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Nicholas J Tursi
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shushu Zhao
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Zev A Binder
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Philadelphia, Pennsylvania, USA
| | - Donald O’Rourke
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Philadelphia, Pennsylvania, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Rossi J, Zedde M, Napoli M, Pascarella R, Pisanello A, Biagini G, Valzania F. Impact of Sex Hormones on Glioblastoma: Sex-Related Differences and Neuroradiological Insights. Life (Basel) 2024; 14:1523. [PMID: 39768232 PMCID: PMC11677825 DOI: 10.3390/life14121523] [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: 10/05/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Glioblastoma (GBM) displays significant gender disparities, being 1.6 times more prevalent in men, with a median survival time of 15.0 months for males compared to 25.5 months for females. These differences may be linked to gonadal steroid hormones, particularly testosterone, which interacts with the androgen receptor (AR) to promote tumor proliferation. Conversely, estrogen (E2), progesterone (P4), and P4 metabolites exert more complex effects on GBM. Despite these insights, the identification of reliable hormonal tumor markers remains challenging, and studies investigating hormone therapies yield inconclusive results due to small sample sizes and heterogeneous tumor histology. Additionally, genetic, epigenetic, and immunological factors play critical roles in sex disparities, with female patients demonstrating increased O6-Methylguanine-DNA methyltransferase promoter methylation and greater genomic instability. These complexities highlight the need for personalized therapeutic strategies that integrate hormonal influences alongside other sex-specific biological characteristics in the management of GBM. In this review, we present the current understanding of the potential role of sex hormones in the natural history of GBM.
Collapse
Affiliation(s)
- Jessica Rossi
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy;
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy (F.V.)
| | - Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy (F.V.)
| | - Manuela Napoli
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy; (M.N.); (R.P.)
| | - Rosario Pascarella
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy; (M.N.); (R.P.)
| | - Anna Pisanello
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy (F.V.)
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Franco Valzania
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy (F.V.)
| |
Collapse
|
23
|
Jo H, Lee S, Kim MH, Park S, Lee SY. Recapitulating Glioma Stem Cell Niches Using 3D Spheroid Models for Glioblastoma Research. BIOSENSORS 2024; 14:539. [PMID: 39589998 PMCID: PMC11592235 DOI: 10.3390/bios14110539] [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: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Glioblastoma multiforme (GBM) is among the most aggressive brain cancers, and it contains glioma stem cells (GSCs) that drive tumor initiation, progression, and recurrence. These cells resist conventional therapies, contributing to high recurrence rates in GBM patients. Developing in vitro models that mimic the tumor microenvironment (TME), particularly the GSC niche, is crucial for understanding GBM growth and therapeutic resistance. Three-dimensional (3D) spheroid models provide a more physiologically relevant approach than traditional two-dimensional (2D) cultures, recapitulating key tumor features like hypoxia, cell heterogeneity, and drug resistance. This review examines scaffold-free and scaffold-based methods for generating 3D GBM spheroids, focusing on their applications in studying the cancer stem cell niche. The discussion encompasses methods such as the hanging drop, low-adhesion plates, and magnetic levitation, alongside advancements in embedding spheroids within extracellular matrix-based hydrogels and employing 3D bioprinting to fabricate more intricate tumor models. These 3D culture systems offer substantial potential for enhancing our understanding of GBM biology and devising more effective targeted therapies.
Collapse
Affiliation(s)
- Hyunji Jo
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (H.J.); (S.L.)
| | - Seulgi Lee
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (H.J.); (S.L.)
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea;
| | - Sungsu Park
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (H.J.); (S.L.)
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea;
- Department of Quantum Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Seo-Yeon Lee
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
- Department of Biomedical Science, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| |
Collapse
|
24
|
Digiovanni S, Lorenzati M, Bianciotto OT, Godel M, Fontana S, Akman M, Costamagna C, Couraud PO, Buffo A, Kopecka J, Riganti C, Salaroglio IC. Blood-brain barrier permeability increases with the differentiation of glioblastoma cells in vitro. Fluids Barriers CNS 2024; 21:89. [PMID: 39487455 PMCID: PMC11529439 DOI: 10.1186/s12987-024-00590-0] [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: 12/31/2023] [Accepted: 10/24/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive tumor, difficult to treat pharmacologically because of the blood-brain barrier (BBB), which is rich in ATP-binding cassette (ABC) transporters and tight junction (TJ) proteins. The BBB is disrupted within GBM bulk, but it is competent in brain-adjacent-to-tumor areas, where eventual GBM foci can trigger tumor relapse. How GBM cells influence the permeability of BBB is poorly investigated. METHODS To clarify this point, we co-cultured human BBB models with 3 patient-derived GBM cells, after separating from each tumor the stem cell/neurosphere (SC/NS) and the differentiated/adherent cell (AC) components. Also, we set up cultures of BBB cells with the conditioned medium of NS or AC, enriched or depleted of IL-6. Extracellular cytokines were measured by protein arrays and ELISA. The intracellular signaling in BBB cells was measured by immunoblotting, in the presence of STAT3 pharmacological inhibitor or specific PROTAC. The competence of BBB was evaluated by permeability assays and TEER measurement. RESULTS The presence of GBM cells or their conditioned medium increased the permeability to doxorubicin, mitoxantrone and dextran-70, decreased TEER, down-regulated ABC transporters and TJ proteins at the transcriptional level. These effects were higher with AC or their medium than with NS. The secretome analysis identified IL-6 as significantly more produced by AC than by NS. Notably, AC-conditioned medium treated with an IL-6 neutralizing antibody reduced the BBB permeability to NS levels, while NS-conditioned medium enriched with IL-6 increased BBB permeability to AC levels. Mechanistically, IL-6 released by AC GBM cells activated STAT3 in BBB cells. In turn, STAT3 down-regulated ABC transporter and TJ expression, increased permeability and decreased TEER. The same effects were obtained in BBB cells treated with STA-21, a pharmacological inhibitor of STAT3, or with a PROTAC targeting STAT3. CONCLUSIONS Our work demonstrates for the first time that the degree of GBM differentiation influences BBB permeability. The crosstalk between GBM cells that release IL-6 and BBB cells that respond by activating STAT3, controls the expression of ABC transporters and TJ proteins on BBB. These results may pave the way for novel therapeutic tools to tune BBB permeability and improve drug delivery to GBM.
Collapse
Affiliation(s)
- Sabrina Digiovanni
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Martina Lorenzati
- Department of Neuroscience Rita Levi Montalcini, Institute Cavalieri Ottolenghi, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Olga Teresa Bianciotto
- Department of Neuroscience Rita Levi Montalcini, Institute Cavalieri Ottolenghi, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Martina Godel
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Simona Fontana
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Muhlis Akman
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Costanzo Costamagna
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | | | - Annalisa Buffo
- Department of Neuroscience Rita Levi Montalcini, Institute Cavalieri Ottolenghi, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy.
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy.
| | - Iris Chiara Salaroglio
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| |
Collapse
|
25
|
Liu Y, Fang S, Wang P, Zhang J, Liu F. Olaparib Enhances the Efficacy of Third-Generation Oncolytic Adenoviruses Against Glioblastoma by Modulating DNA Damage Response and p66shc-Induced Apoptosis. CNS Neurosci Ther 2024; 30:e70124. [PMID: 39552450 PMCID: PMC11570871 DOI: 10.1111/cns.70124] [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: 07/22/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024] Open
Abstract
AIMS Patients with glioblastoma multiforme (GBM) do not benefit from current cancer treatments, and their prognosis is dismal. This study aimed to investigate the potential synergistic effects of TS-2021, a third-generation oncolytic adenovirus, combined with the PARP inhibitor olaparib in GBM. METHODS TS-2021's impact on p66shc-induced apoptosis, DNA damage response, and poly (ADP-ribose) polymerase (PARP) activation was evaluated in GBM cells. The synergistic effect of TS-2021 and olaparib was examined in GBM cell lines and an immunocompetent mouse model of GBM. Mechanistic studies focused on the role of p66shc phosphorylation in the observed effects. RESULTS TS-2021 triggered p66shc-induced apoptosis, DNA damage response, and PARP activation. The combination of TS-2021 and olaparib synergistically increased cell apoptosis and DNA damage and reduced PARP expression compared to monotherapy. Olaparib promoted TS-2021 replication and release in GBM cells. Mechanistically, olaparib combined with TS-2021 upregulated p66shc phosphorylation, enhancing tumor cell apoptosis. In vivo, the combination therapy inhibited tumor growth and prolonged survival, confirming the synergistic effect. CONCLUSION This study is the first to suggest that TS-2021 sensitizes GBM cells with wild-type BRCA1/2 to olaparib. The combination of TS-2021 and olaparib shows a synergistic therapeutic effect against GBM, providing a promising treatment strategy.
Collapse
Affiliation(s)
- Yida Liu
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Peiwen Wang
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| |
Collapse
|
26
|
Jiang C, Wu W, Jiang X, Qian K. Integrative multi-omics analysis unveils the connection between transcriptomic characteristics associated with mitochondria and the tumor immune microenvironment in lower-grade gliomas. Sci Rep 2024; 14:23675. [PMID: 39390013 PMCID: PMC11467307 DOI: 10.1038/s41598-024-74281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
Lower-grade gliomas (LGGs) exhibit diverse clinical behaviors and varying immune infiltration levels. Mitochondria have been implicated in numerous cancer pathogenesis and development, including LGGs. However, the precise biological functions of mitochondrial genes in shaping the immune landscape and the prognostic significance of LGGs remain elusive. Utilizing the Mito-Carta3.0 database, we curated a total of 1136 genes implicated in mitochondrial functions. By leveraging the expression profiles of 1136 genes related to mitochondria, we successfully categorized LGGs into four distinctive mitochondria-related transcriptome (MRT) subtypes. Our thorough analysis conclusively demonstrated that these subtypes exhibited marked disparities. To enable a personalized and integrated evaluation of LGG patients, we developed a prognostic signature known as MRT-related prognostic signature (MTRS). MTRS demonstrated correlation with mitochondria-related transcriptome (MRT) subtypes, allowing the assessment of patients' prognosis and immune microenvironment. We conducted a detailed exploration of the single-cell distribution of MTRS in lower-grade gliomas and verified the core genes of MTRS within the spatial transcriptome of these tumors. Furthermore, our study pinpointed MGME1 as the pivotal gene in the model, functioning as an oncogene that exerts influence on cell proliferation and migration capabilities. Our research highlights the importance of mitochondrial transcriptomic features in LGGs, offering paths for tailored therapies.
Collapse
Affiliation(s)
- Cheng Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Wenjie Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Kang Qian
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
27
|
Chuang SH, Chen KJ, Cheng YT, Chen YS, Lin SY, Chou HY, Tsai HC. A thermo-responsive chemically crosslinked long-term-release chitosan hydrogel system increases the efficiency of synergy chemo-immunotherapy in treating brain tumors. Int J Biol Macromol 2024; 280:135894. [PMID: 39322160 DOI: 10.1016/j.ijbiomac.2024.135894] [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: 04/29/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Glioblastoma multiforme (GBM) is an aggressive and common brain tumor. The blood-brain barrier prevents several treatments from reaching the tumor. This study proposes a Chemo-Immunotherapy synergy treatment chemically crosslinked hydrogel system that is injected into the tumor to treat GBM. The strategy uses doxorubicin and BMS-1 with a thermo-responsive and chemically crosslinked hydrogel for extended drug release into the affected area. The hydrogels are produced by mixing with Chitosan (Chi), modified Pluronic F-127 (PF-127) with aldehyde end group and doxorubicin and then chemically crosslinking the aldehyde and amine bonds to increase the drug retention time. PF-127-CHO/Chi, which gels at body temperatures and chemically crosslinks between PF-127-CHO and Chitosan, increases the time that the drug remains in the affected area and prevents the hydrogel from swelling and compressing surrounding tissue. The drug is released from the chemically crosslinked hydrogels, prevents tumor progression and increases survival for subjects with GBM tumors. The Synergy Chemo-Immunotherapy also allows more efficient treatment of GBM than chemotherapy. The PF-127-CHO/Chi DOX and BMS-1 group have a tumor that is 43 times smaller than the untreated group. These results show that the proposed chemically crosslinking hydrogel is an efficient intratumoral delivery platform for the treatment of tumors.
Collapse
Affiliation(s)
- Shun-Hao Chuang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Kuan-Ju Chen
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Yu-Ting Cheng
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Yu-Shuan Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Taiwan; Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
| | - Shuian-Yin Lin
- Biomedical Technology and Device Research Center, Industrial Technology Research Institute, Hsinchu, 310, Taiwan
| | - Hsiao-Ying Chou
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei 106, Taiwan; R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan.
| |
Collapse
|
28
|
Tanriverdi G, Kaleci B, Yavuz F, Sahin H, Purelku M, Yazici Z, Kokturk S. The effects of the combination of temozolomide and Eribulin on T98G human glioblastoma cell line: an ultrastructural study. Ultrastruct Pathol 2024; 48:323-337. [PMID: 38916264 DOI: 10.1080/01913123.2024.2371821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
Glioblastoma tumors are the most aggressive primary brain tumors that develop resistance to temozolomide (TMZ). Eribulin (ERB) exhibits a unique mechanism of action by inhibiting microtubule dynamics during the G2/M cell cycle phase. We utilized the T98G human glioma cell line to investigate the effects of ERB and TMZ, both individually and in combination. The experimental groups were established as follows: control, E5 (5 nM ERB), T0.75 (0.75 mM TMZ), T1 (1.0 mM TMZ), and combination groups (E5+T0.75 and E5+T1). All groups showed a significant decrease in cell proliferation. Apoptotic markers revealed a time-dependent increase in annexin-V expression, across all treatment groups at the 48-hour time point. Caspase-3, exhibited an increase in the combination treatment groups at the 48-hour mark. Transmission electron microscopy (TEM) revealed normal ultrastructural features in the glioma cells of the control group. However, treatments induced ultrastructural changes within the spheroid glioblastoma model, particularly in the combination groups. These changes included a dose-dependent increase in autophagic vacuoles and apoptotic morphology of the cells. In conclusion, the similarity in the mechanism of action between ERB and TMZ suggests the potential for synergistic effects when combined. Our results highlight that this combination induced severe damage and autophagy in glioma spheroids after 48 hours.
Collapse
Affiliation(s)
- Gamze Tanriverdi
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Belisa Kaleci
- Ministry of Health and Social Protection, University Dental Clinic, Tirane, Albania
| | - Furkan Yavuz
- Radiation Oncology Department, School of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Hakan Sahin
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Merjem Purelku
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeliha Yazici
- Medical Pharmacology, Medicine, Istanbul Arel University, İstanbul, Türkiye
| | - Sibel Kokturk
- Department of Histology and Embryology, Medicine Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
29
|
Dandagi P, K YB, Mary Martin T, K MS. In Silico and Glioblastoma Cell Line Evaluation of Thioflavin-Derived Zinc Nanoparticles Targeting Beclin Protein. Cureus 2024; 16:e69319. [PMID: 39398744 PMCID: PMC11471010 DOI: 10.7759/cureus.69319] [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: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
INTRODUCTION This study explores the anticancer potential of Thioflavin-derived zinc nanoparticles (Th-ZnNPs) using both in vitro and in silico methods. Thioflavin, known for its specific binding properties, faces challenges such as bioavailability, rapid metabolism, and solubility. To overcome these limitations and enhance therapeutic efficacy, nanotechnology was utilized to synthesize Th-ZnNPs. These nanoparticles (NPs) are designed to improve drug delivery and effectiveness. The Beclin protein, which plays a critical role in regulating autophagy in cancer cells, was identified as a potential target for these NPs. The study aims to evaluate the interaction between Th-ZnNPs and Beclin protein in glioblastoma cell lines and assess the potential of these NPs as novel anticancer agents. METHODS Th-ZnNPs were synthesized using advanced nanotechnology techniques to improve the bioavailability and solubility of Thioflavin. To explore their anticancer potential, in silico analyses were performed, including molecular docking studies to evaluate the binding affinity between the ZnNPs and Beclin protein, which is integral to autophagy regulation. This computational approach identified the Beclin protein as a promising target for the ZnNPs. Complementary in vitro assays were then conducted, where glioblastoma cell lines (procured from the National Centre for Cell Science, Pune, India) were treated with ZnNPs to assess their cytotoxic effects. The assays also included mechanistic studies to validate the interaction between ZnNPs and Beclin protein and to understand their influence on autophagy pathways. RESULTS The synthesis of Th-ZnNPs successfully enhanced their solubility and bioavailability compared to Thioflavin alone. In silico findings showed a strong binding affinity between the Th-ZnNPs and the Beclin protein, suggesting that these NPs may effectively target cancer cells through this interaction. Beclin protein was validated as a relevant target due to its critical role in autophagy regulation. In vitro assays further confirmed the anticancer potential of the Th-ZnNPs, as they exhibited significant cytotoxic effects on glioblastoma cells. Additionally, mechanistic studies revealed that Th-ZnNPs impact Beclin protein and modulate autophagy pathways, supporting their proposed role as effective anticancer agents. CONCLUSIONS The study highlights the promising anticancer potential of Th-ZnNPs. By overcoming the limitations of Thioflavin through nanotechnology, these NPs show significant therapeutic promise in targeting glioblastoma cells. The strong binding affinity between Th-ZnNPs and the Beclin protein, coupled with confirmed cytotoxic effects, underscores their potential as novel anticancer agents. This integrated approach not only enhances the delivery and efficacy of Thioflavin but also opens new avenues for targeted therapy in glioblastoma treatment.
Collapse
Affiliation(s)
- Parineeta Dandagi
- Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Yuvaraj Babu K
- Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Taniya Mary Martin
- Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Meenakshi Sundaram K
- Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
30
|
Bahojb Mahdavi SZ, Pouladi N, Amini M, Baradaran B, Najafi S, Vaghef Mehrabani S, Yari A, Ghobadi Alamdari S, Mokhtarzadeh AA. Let-7a-3p overexpression increases chemosensitivity to carmustine and synergistically promotes autophagy and suppresses cell survival in U87MG glioblastoma cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6903-6918. [PMID: 38587542 DOI: 10.1007/s00210-024-03060-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
In terms of primary brain tumors, glioblastoma is one of the most aggressive and common brain tumors. The high resistance of glioblastoma to chemotherapy has made it vital to find alternative treatments and biological mechanisms to reduce the survival of cancer cells. Given that, the objective of the present research was to explore the potential of let-7a-3p when used in combination with carmustine in human glioblastoma cancer cells. Based on previous studies, the expression of let-7a is downregulated in the U87MG cell line. Let-7a-3p transfected into U87MG glioblastoma cells. Cell viability of the cells was assessed by MTT assay. The apoptotic induction in U87MG cancerous cells was determined through the utilization of DAPI and Annexin V/PI staining techniques. Moreover, the induction of autophagy and cell cycle arrest was evaluated by flow cytometry. Furthermore, cell migration was evaluated by the wound healing assay while colony formation assay was conducted to evaluate colony formation. Also, the expression of the relevant genes was evaluated using qRT-PCR. Transfection of let-7a-3p mimic in U87MG cells increased the expression of the miRNA and also increased the sensitivity of U87MG cells to carmustine. Let-7a-3p and carmustine induced sub-G1 and S phase cell cycle arrest, respectively. Combination treatment of let-7a-3p and carmustine synergistically increased arrested cells and induced apoptosis through regulating involved genes including P53, caspase-3, Bcl-2, and Bax. Combined treatment with let-7a-3p and carmustine also induced autophagy and increased the expression of the ATG5 and Beclin 1 (ATG6). Furthermore, let-7a-3p combined with carmustine inhibited cell migration via decreasing the expression of MMP-2. Moreover, the combination therapy decreased the ability of U87MG to form colonies through downregulating CD-44. In conclusion, our work suggests that combining let-7a-3p replacement therapy with carmustine treatment could be considered a promising strategy in treatment and can increase efficiency of glioblastoma chemotherapy.
Collapse
Affiliation(s)
- Seyedeh Zahra Bahojb Mahdavi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Vaghef Mehrabani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sania Ghobadi Alamdari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, Iran
| | | |
Collapse
|
31
|
McBriar JD, Shafiian N, Scharf S, Boockvar JA, Wernicke AG. Prostate-Specific Membrane Antigen Use in Glioma Management: Past, Present, and Future. Clin Nucl Med 2024; 49:806-816. [PMID: 38968568 DOI: 10.1097/rlu.0000000000005365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) is a membrane-bound metallopeptidase highly expressed in the neovasculature of many solid tumors including gliomas. It is a particularly enticing therapeutic target due to its ability to internalize, thereby delivering radioligands or pharmaceuticals to the intracellular compartment. Targeting the neovasculature of gliomas using PSMA for diagnosis and management has been a recent area of increased study and promise. The purpose of this review is to synthesize the current state and future directions of PSMA use in the histopathologic study, imaging, and treatment of gliomas. METHODS PubMed and Scopus databases were used to conduct a literature review on PSMA use in gliomas in June 2023. Terms searched included "PSMA," "Prostate-Specific Membrane Antigen" OR "PSMA" OR "PSMA PET" AND "glioma" OR "high grade glioma" OR "glioblastoma" OR "GBM." RESULTS Ninety-four publications were screened for relevance with 61 studies, case reports, and reviews being read to provide comprehensive context for the historical, contemporary, and prospective use of PSMA in glioma management. CONCLUSIONS PSMA PET imaging is currently a promising and accurate radiographic tool for the diagnosis and management of gliomas. PSMA histopathology likely represents a viable tool for helping predict glioma behavior. More studies are needed to investigate the role of PSMA-targeted therapeutics in glioma management, but preliminary reports have indicated its potential usefulness in treatment.
Collapse
Affiliation(s)
- Joshua D McBriar
- From the Donald and Barbara Zucker School of Medicine at Hofstra/Northwell
| | - Neeva Shafiian
- From the Donald and Barbara Zucker School of Medicine at Hofstra/Northwell
| | | | | | - A Gabriella Wernicke
- Radiation Medicine, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY
| |
Collapse
|
32
|
Yoo S, Kim M, Choi C, Kim DH, Cha GD. Soft Bioelectronics for Neuroengineering: New Horizons in the Treatment of Brain Tumor and Epilepsy. Adv Healthc Mater 2024; 13:e2303563. [PMID: 38117136 DOI: 10.1002/adhm.202303563] [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: 10/17/2023] [Revised: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Soft bioelectronic technologies for neuroengineering have shown remarkable progress, which include novel soft material technologies and device design strategies. Such technological advances that are initiated from fundamental brain science are applied to clinical neuroscience and provided meaningful promises for significant improvement in the diagnosis efficiency and therapeutic efficacy of various brain diseases recently. System-level integration strategies in consideration of specific disease circumstances can enhance treatment effects further. Here, recent advances in soft implantable bioelectronics for neuroengineering, focusing on materials and device designs optimized for the treatment of intracranial disease environments, are reviewed. Various types of soft bioelectronics for neuroengineering are categorized and exemplified first, and then details for the sensing and stimulating device components are explained. Next, application examples of soft implantable bioelectronics to clinical neuroscience, particularly focusing on the treatment of brain tumor and epilepsy are reviewed. Finally, an ideal system of soft intracranial bioelectronics such as closed-loop-type fully-integrated systems is presented, and the remaining challenges for their clinical translation are discussed.
Collapse
Affiliation(s)
- Seungwon Yoo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Minjeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Changsoon Choi
- Center for Opto-Electronic Materials and Devices, Post-silicon Semiconductor Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Gi Doo Cha
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| |
Collapse
|
33
|
Zheng G, Zeng G, Wei D. The role of NPC2 gene in glioma was investigated based on bioinformatics analysis. Sci Rep 2024; 14:19134. [PMID: 39160329 PMCID: PMC11333723 DOI: 10.1038/s41598-024-70221-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/14/2024] [Indexed: 08/21/2024] Open
Abstract
Glioblastoma (GBM) is one of the most malignant primary brain tumors in adults. The NPC2 gene (Niemann-Pick type C intracellular cholesterol transporter 2) is a protein-coding gene with a lipid recognition domain. The NPC2 gene was found to be significantly increased in gliomas (LGG and GBM), and it is now thought to be a risk factor. COX analysis demonstrated that NPC2 was a significant risk factor for glioma. Functional enrichment analysis of genes that were differentially expressed between high and low expression groups revealed that genes were primarily enriched in the regulation of trans-synaptic signaling, Retrograde endocannabinoid signaling and other pathways. According to the findings of the immunoinfiltration investigation, the NPC2 gene and macrophage, DC, etc. have a strong positive association. In addition, patients with high NPC2 expression had higher levels of immune cell expression. Medication sensitivity research revealed that NPC2's differential expression had some bearing on patients' medication sensitivity. There was a strong correlation between the prognosis of glioma patients and the gene sets NUDT19 and NUME. In brief, the NPC2 gene was identified to be a possible biomarker of glioma, and preliminary analysis was done on the role of the NPC2 gene in immunological microenvironment of glioma.
Collapse
Affiliation(s)
- Guangwei Zheng
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Guangming Zeng
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - De Wei
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
34
|
Song B, Wang X, Qin L, Hussain S, Liang W. Brain gliomas: Diagnostic and therapeutic issues and the prospects of drug-targeted nano-delivery technology. Pharmacol Res 2024; 206:107308. [PMID: 39019336 DOI: 10.1016/j.phrs.2024.107308] [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: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Glioma is the most common intracranial malignant tumor, with severe difficulty in treatment and a low patient survival rate. Due to the heterogeneity and invasiveness of tumors, lack of personalized clinical treatment design, and physiological barriers, it is often difficult to accurately distinguish gliomas, which dramatically affects the subsequent diagnosis, imaging treatment, and prognosis. Fortunately, nano-delivery systems have demonstrated unprecedented capabilities in diagnosing and treating gliomas in recent years. They have been modified and surface modified to efficiently traverse BBB/BBTB, target lesion sites, and intelligently release therapeutic or contrast agents, thereby achieving precise imaging and treatment. In this review, we focus on nano-delivery systems. Firstly, we provide an overview of the standard and emerging diagnostic and treatment technologies for glioma in clinical practice. After induction and analysis, we focus on summarizing the delivery methods of drug delivery systems, the design of nanoparticles, and their new advances in glioma imaging and treatment in recent years. Finally, we discussed the prospects and potential challenges of drug-delivery systems in diagnosing and treating glioma.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Shehbaz Hussain
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
35
|
Cai X, Qian M, Zhang K, Li Y, Chang B, Chen M. Profiling and Bioinformatics Analyses of Hypoxia-Induced Differential Expression of Long Non-coding RNA in Glioblastoma Multiforme Cells. Biochem Genet 2024; 62:3052-3070. [PMID: 38066404 DOI: 10.1007/s10528-023-10597-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/08/2023] [Indexed: 07/31/2024]
Abstract
Hypoxic microenvironments are intricately linked to malignant characteristics of glioblastoma multiforme (GBM). Long non-coding ribonucleic acids (lncRNAs) have been reported to be involved in the progression of GBM and closely associated with hypoxia. Nevertheless, the differential expression profiles as well as functional roles of lncRNAs in GBM cells under hypoxic conditions remain largely obscure. We explored the expression profiles of lncRNAs in hypoxic U87 cells as well as T98G cells using sequencing analysis. The effect of differentially expressed lncRNAs (DElncRNAs) was assessed through bioinformatic analysis. Furthermore, the expression of lncRNAs significantly dysregulated in both U87 and T98G cells was further validated using quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Relevant cell functional experiments were also conducted. We used predicted RNA-binding proteins (RBPs) to construct an interaction network via the interaction prediction module. U87 and T98G cells showed dysregulation of 1115 and 597 lncRNAs, respectively. Gene Ontology (GO) analysis indicated that altered lncRNA expression was associated with nucleotide-excision repair and cell metabolism in GBM cells. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed the association between dysregulated lncRNAs and the Hippo signaling pathway under hypoxia. The dysregulation of six selected lncRNAs (ENST00000371192, uc003tnq.3, ENST00000262952, ENST00000609350, ENST00000610036, and NR_046262) was validated by qRT-PCR. Investigation of lncRNA-microRNA (miRNA)-mRNA networks centered on HIF-1α demonstrated cross-talk between the six validated lncRNAs and 16 related miRNAs. Functional experiments showed the significant inhibition of GBM cell proliferation, invasion, and migration by the knockdown of uc003tnq.3 in vitro. Additionally, uc003tnq.3 was used to construct a comprehensive RBP-transcription factor (TF)-miRNA interaction network. The expression of LncRNAs was dysregulated in GBM cells under hypoxic conditions. The identified six lncRNAs might exert important effect on the development of GBM under hypoxic microenvironment.
Collapse
Affiliation(s)
- Xiaomin Cai
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Yangpu District, Shanghai, 200092, China
| | - Mengshu Qian
- Department of Emergency and Critical Care Medicine, Kong Jiang Hospital of Yangpu District, Shanghai, 200082, China
| | - Kui Zhang
- Department of Plastic Surgery, Xuzhou Medical University Affiliated Xuzhou City Hospital, Xuzhou, 221000, Jiangsu, China
| | - Yanzhen Li
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Yangpu District, Shanghai, 200092, China
| | - Bowen Chang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lu Jiang Road, Luyang District, Hefei, 230001, Anhui, China.
| | - Ming Chen
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
36
|
Seyhan AA. Circulating Liquid Biopsy Biomarkers in Glioblastoma: Advances and Challenges. Int J Mol Sci 2024; 25:7974. [PMID: 39063215 PMCID: PMC11277426 DOI: 10.3390/ijms25147974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Gliomas, particularly glioblastoma (GBM), represent the most prevalent and aggressive tumors of the central nervous system (CNS). Despite recent treatment advancements, patient survival rates remain low. The diagnosis of GBM traditionally relies on neuroimaging methods such as magnetic resonance imaging (MRI) or computed tomography (CT) scans and postoperative confirmation via histopathological and molecular analysis. Imaging techniques struggle to differentiate between tumor progression and treatment-related changes, leading to potential misinterpretation and treatment delays. Similarly, tissue biopsies, while informative, are invasive and not suitable for monitoring ongoing treatments. These challenges have led to the emergence of liquid biopsy, particularly through blood samples, as a promising alternative for GBM diagnosis and monitoring. Presently, blood and cerebrospinal fluid (CSF) sampling offers a minimally invasive means of obtaining tumor-related information to guide therapy. The idea that blood or any biofluid tests can be used to screen many cancer types has huge potential. Tumors release various components into the bloodstream or other biofluids, including cell-free nucleic acids such as microRNAs (miRNAs), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), proteins, extracellular vesicles (EVs) or exosomes, metabolites, and other factors. These factors have been shown to cross the blood-brain barrier (BBB), presenting an opportunity for the minimally invasive monitoring of GBM as well as for the real-time assessment of distinct genetic, epigenetic, transcriptomic, proteomic, and metabolomic changes associated with brain tumors. Despite their potential, the clinical utility of liquid biopsy-based circulating biomarkers is somewhat constrained by limitations such as the absence of standardized methodologies for blood or CSF collection, analyte extraction, analysis methods, and small cohort sizes. Additionally, tissue biopsies offer more precise insights into tumor morphology and the microenvironment. Therefore, the objective of a liquid biopsy should be to complement and enhance the diagnostic accuracy and monitoring of GBM patients by providing additional information alongside traditional tissue biopsies. Moreover, utilizing a combination of diverse biomarker types may enhance clinical effectiveness compared to solely relying on one biomarker category, potentially improving diagnostic sensitivity and specificity and addressing some of the existing limitations associated with liquid biomarkers for GBM. This review presents an overview of the latest research on circulating biomarkers found in GBM blood or CSF samples, discusses their potential as diagnostic, predictive, and prognostic indicators, and discusses associated challenges and future perspectives.
Collapse
Affiliation(s)
- Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
37
|
Coupey J, Leblond MM, Hue ES, Valable S. Flow cytometry detection and quantification of circulating leukocyte subpopulations in mice after brain irradiation. Methods Cell Biol 2024; 189:135-152. [PMID: 39393880 DOI: 10.1016/bs.mcb.2024.06.004] [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: 10/13/2024]
Abstract
In the context of high-grade gliomas such as glioblastoma (GBM), the immune part of the tumor microenvironment (TME) is involved in tumor growth and tumor recurrence. It is mostly represented by high amount of macrophages and low amount of lymphocytes. GBM in itself as well as x-ray-based radiotherapy, a standard treatment for brain tumors, are also associated with systemic effects like lymphopenia that correlates with a poor prognosis. This contributes to the immune-suppressive nature of the TME and may explain the lack of the anti-tumor immune response. Radiation-induced lymphopenia (RIL) is generally evaluated on CD4+ and CD8+ count or on a CBC (complete blood count), but the heterogeneity of the subtypes prompts us to explore them in detail to better understand the cellular response to brain irradiation. To facilitate and develop the evaluation of x-ray brain exposure on circulating immune cells, we developed a reproducible and reliable method to quantify the variation of lymphoid and myeloid subtypes using flow cytometry after brain irradiation in the rodent.
Collapse
Affiliation(s)
- Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France
| | - Marine M Leblond
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Erika S Hue
- LABÉO, Saint-Contest, France; Normandie Université, UNICAEN BIOTARGEN, Saint-Contest, France
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France.
| |
Collapse
|
38
|
Zhao K, Xu J, Zhao B. Panoramic RNA expression of fibroblast growth factors in human glioblastoma tissues and the impact on the survival of patients. Oncol Lett 2024; 28:317. [PMID: 38807663 PMCID: PMC11130607 DOI: 10.3892/ol.2024.14450] [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: 12/29/2023] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
Fibroblast growth factors (FGFs) have a key role in various critical steps of tumor growth and progression through effects on angiogenesis, inflammation and the growth and invasion of malignant cells. Nevertheless, the role of the FGF family in human glioblastoma (GBM) has been rarely studied. The objective of the present study was to assess the RNA expression of all FGF family members in tissues obtained from patients with GBM and to analyze the association between FGF expression and the survival of these patients. For this, the RNA expression of FGF family members in the malignant and proximal tissues of 12 patients with GBM was determined by analyzing high-throughput RNA transcriptome sequencing data uploaded to the National Center for Biotechnology Information database. The relationship between FGF genes and the survival of patients with GBM and glioma was also respectively studied by analyzing data from The Cancer Genome Atlas database using the Gene Expression Profiling Interactive Analysis tool. The results showed that the expression of FGF1, FGF17, FGF20 and FGF22 in GBM tissues was lower than that in adjacent tissues, with a difference of >2 times. Analysis of the overall survival of patients with GBM indicated there were no significant relationships between the expression of FGF1, FGF17, FGF20, FGF22 and overall survival. Analysis of the overall survival of patients with glioma showed that glioma patients with low FGF1 expression achieved a longer survival time than patients with high FGF1 expression; however, high expression of FGF17 and FGF22 indicated a longer survival time. In summary, the results of the present study demonstrated the panoramic expression of FGF family members in patients with GBM, and indicated that FGF1, FGF17 and FGF22 did not affect the survival of patients with GBM, but had a notable influence on the survival of patients with glioma.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jiakun Xu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Beichuan Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
39
|
Gersey ZC, Plute T, Jaman E, Zhang X, Mitha R, Zinn PO, Pearce TM, Amankulor NM. Cerebrospinal Fluid Seeding Versus Inflammation in Setting of Ventriculoperitoneal Shunt as a Potential Cause for Distant Recurrence of Glioblastoma. Brain Tumor Res Treat 2024; 12:181-185. [PMID: 39109619 PMCID: PMC11306840 DOI: 10.14791/btrt.2024.0015] [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: 03/28/2024] [Revised: 05/16/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults with a median survival of approximately 15 months, despite treatment, with most patients experiencing recurrence within 9 months of resection. The propensity of recurrence in GBM exemplifies the fatal course of the disease and remains an underlying area of study as novel instances of recurrence are encountered. The authors present a unique case of a 31-year-old male patient with a history of cerebellomedullary junction astrocytoma who later developed a supratentorial GBM followed by recurrence centered around a preexisting ventriculoperitoneal catheter and located in the hemisphere contralateral to his first GBM. Each of these lesions was initially thought to represent de novo glial neoplasms because of the absence of intervening T2 fluid-attenuated inversion recovery signal change between each lesion. However, next-generation sequencing using the GlioSeq™ platform revealed similar mutational profiles in both GBMs, suggesting an alternative method of migration of tumor cells to the shunt catheter site, and a local inflammatory environment likely triggering recurrence. This study concludes that in rare instances, in the presence of dormant glioma cells, intracranial foreign bodies may promote an inflammatory microenvironment that may activate tumorigenesis.
Collapse
Affiliation(s)
- Zachary C Gersey
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Tritan Plute
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Emade Jaman
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Xiaoran Zhang
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rida Mitha
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Pascal O Zinn
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Thomas M Pearce
- Division of Anatomic Pathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nduka M Amankulor
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
40
|
Hao W, Sun N, Fan Y, Chen M, Liu Q, Yang M, Yang Y, Gao C. Targeted Ferroptosis-Immunotherapy Synergy: Enhanced Antiglioma Efficacy with Hybrid Nanovesicles Comprising NK Cell-Derived Exosomes and RSL3-Loaded Liposomes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28193-28208. [PMID: 38776411 PMCID: PMC11164066 DOI: 10.1021/acsami.4c04604] [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: 03/20/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Ferroptosis therapy and immunotherapy have been widely used in cancer treatment. However, nonselective induction of ferroptosis in tumors is prone to immunosuppression, limiting the therapeutic effect of ferroptosis cancer treatment. To address this issue, this study reports a customized hybrid nanovesicle composed of NK cell-derived extracellular versicles and RSL3-loaded liposomes (hNRVs), aiming to establish a positive cycle between ferroptosis therapy and immunotherapy. Thanks to the enhanced permeability and retention effect and the tumor homing characteristics of NK exosomes, our data indicate that hNRVs can actively accumulate in tumors and enhance cellular uptake. FASL, IFN-γ, and RSL3 are released into the tumor microenvironment, where FASL derived from NK cells effectively lyses tumor cells. RSL3 downregulates the expression of GPX4 in the tumor, leading to the accumulation of LPO and ROS, and promotes ferroptosis in tumor cells. The accumulation of IFN-γ and TNF-α stimulates the maturation of dendritic cells and effectively induces the inactivation of GPX4, promoting lipid peroxidation, making them sensitive to ferroptosis and indirectly promoting the occurrence of ferroptosis. This study highlights the role of the customized hNRV platform in enhancing the effectiveness of synergistic treatment with selective delivery of ferroptosis inducers and immune activation against glioma without causing additional side effects on healthy organs.
Collapse
Affiliation(s)
- Wenyan Hao
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| | - Nan Sun
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| | - Yueyue Fan
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| | - Mengyu Chen
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| | - Qianqian Liu
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| | - Meiyan Yang
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Yang
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunsheng Gao
- State Key Laboratory
of Toxicology
and Medical Countermeasures, Beijing Institute
of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
41
|
Onciul R, Toader C, Glavan LA, Covache-Busuioc RA, Bratu BG, Costin HP, Corlatescu AD, Ciurea AV, Grama M, Idu AA. Retrospective Analysis of Glioblastoma Outcomes. Cureus 2024; 16:e62462. [PMID: 38882229 PMCID: PMC11180423 DOI: 10.7759/cureus.62462] [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] [Accepted: 06/15/2024] [Indexed: 06/18/2024] Open
Abstract
This retrospective mono-center study focuses on 144 cases of glioblastoma treated over a time span of 12 years in our clinic in Romania. We offer critical insight into the dreadful aspect of this tumor by highlighting the principal characteristics such as localization, the genetic information of each case, progression-free survival (PFS), and overall survival (OS). A tenth of our patients underwent a second surgical procedure, providing a comparable OS to the other part of our study group, proving that surgical treatment as salvage therapy is a viable option. Also, our research reinforces the fact that utilizing the Karnofsky Performance Scale is a great predictor of patient outcomes in glioblastoma patients. Even though radiotherapy and chemotherapy have mild effects in the context of this oncological disease, our research shows that O6-methylguanine-DNA methyltransferase (MGMT) methylation status and epidermal growth factor receptor (EGFR) amplification have an important effect on OS. Moreover, the particularity of our study, that our patients did not start adjuvant therapy right after surgery, highlighted by a low OS compared to the international literature, sheds light on the fact that chemotherapy and radiotherapy must be started right after the surgical procedure, according to the Stupp protocol. To sum up, our research takes into consideration the factors that influence patient survival and outcome in the battle against glioblastoma.
Collapse
Affiliation(s)
- Razvan Onciul
- Department of Neurosurgery, University Emergency Hospital of Bucharest, Bucharest, ROU
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Corneliu Toader
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, Bucharest, ROU
| | - Luca-Andrei Glavan
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | | | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Horia-Petre Costin
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | | | - Alexandru Vladimir Ciurea
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- Department of Neurosurgery, Sanador Clinical Hospital, Bucharest, ROU
| | - Matei Grama
- Department of Software, Syndical.io, Bucharest, ROU
| | - Andreea-Anamaria Idu
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| |
Collapse
|
42
|
Benkő BM, Tóth G, Moldvai D, Kádár S, Szabó E, Szabó ZI, Kraszni M, Szente L, Fiser B, Sebestyén A, Zelkó R, Sebe I. Cyclodextrin encapsulation enabling the anticancer repositioning of disulfiram: Preparation, analytical and in vitro biological characterization of the inclusion complexes. Int J Pharm 2024; 657:124187. [PMID: 38697585 DOI: 10.1016/j.ijpharm.2024.124187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Drug repositioning is a high-priority and feasible strategy in the field of oncology research, where the unmet medical needs are continuously unbalanced. Disulfiram is a potential non-chemotherapeutic, adjuvant anticancer agent. However, the clinical translation is limited by the drug's poor bioavailability. Therefore, the molecular encapsulation of disulfiram with cyclodextrins is evaluated to enhance the solubility and stability of the drug. The present work describes for the first time the complexation of disulfiram with randomly methylated-β-cyclodextrin. A parallel analytical andin vitrobiological comparison of disulfiram inclusion complexes with hydroxypropyl-β-cyclodextrin, randomly methylated-β-cyclodextrin and sulfobutylether-β-cyclodextrin is conducted. A significant drug solubility enhancement by about 1000-folds and fast dissolution in 1 min is demonstrated. Thein vitrodissolution-permeation studies and proliferation assays demonstrate the solubility-dependent efficacy of the drug. Throughout the different cancer cell lines' characteristics and disulfiram unspecific antitumoral activity, the inhibitory efficacy of the cyclodextrin encapsulated drug on melanoma (IC50 about 100 nM) and on glioblastoma (IC50 about 7000 nM) cell lines differ by a magnitude. This pre-formulation screening experiment serves as a proof of concept of using cyclodextrin encapsulation as a platform tool for further drug delivery development in repositioning areas.
Collapse
Affiliation(s)
- Beáta-Mária Benkő
- University Pharmacy Department of Pharmacy Administration, Semmelweis University, Hőgyes Endre Str. 7-9., Budapest 1092, Hungary.
| | - Gergő Tóth
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre Str. 7-9., Budapest 1092, Hungary.
| | - Dorottya Moldvai
- Tumor Biology, Cell and Tissue Culture Laboratory, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26., Budapest 1085, Hungary.
| | - Szabina Kádár
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre Str. 7-9., Budapest 1092, Hungary; Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest 1111, Hungary.
| | - Edina Szabó
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest 1111, Hungary.
| | - Zoltán-István Szabó
- Faculty of Pharmacy Department of Drugs Industry and Pharmaceutical Management, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Gheorghe Marinescu Str. 38, Târgu Mureș 540142, Romania.
| | - Márta Kraszni
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre Str. 7-9., Budapest 1092, Hungary.
| | - Lajos Szente
- CycloLab Cyclodextrin Research & Development Laboratory Ltd., Illatos út 7, Budapest 1097, Hungary.
| | - Béla Fiser
- Institute of Chemistry, Faculty of Materials Science and Chemical Engineering, University of Miskolc, Egyetemváros, Miskolc 3515, Hungary; Department of Physical Chemistry, Faculty of Chemistry, University of Lodz, 90-236 Lodz, Poland; Ferenc Rakoczi II Transcarpathian Hungarian College of Higher Education, 90200 Beregszász, Transcarpathia, Ukraine.
| | - Anna Sebestyén
- Tumor Biology, Cell and Tissue Culture Laboratory, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26., Budapest 1085, Hungary.
| | - Romána Zelkó
- University Pharmacy Department of Pharmacy Administration, Semmelweis University, Hőgyes Endre Str. 7-9., Budapest 1092, Hungary.
| | - István Sebe
- University Pharmacy Department of Pharmacy Administration, Semmelweis University, Hőgyes Endre Str. 7-9., Budapest 1092, Hungary; Egis Pharmaceuticals Plc., R&D Directorate, P.O. Box 100, Budapest 1475, Hungary.
| |
Collapse
|
43
|
Kostka K, Sokolova V, El-Taibany A, Kruse B, Porada D, Wolff N, Prymak O, Seeds MC, Epple M, Atala AJ. The Application of Ultrasmall Gold Nanoparticles (2 nm) Functionalized with Doxorubicin in Three-Dimensional Normal and Glioblastoma Organoid Models of the Blood-Brain Barrier. Molecules 2024; 29:2469. [PMID: 38893345 PMCID: PMC11173746 DOI: 10.3390/molecules29112469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/28/2024] [Accepted: 05/01/2024] [Indexed: 06/21/2024] Open
Abstract
Among brain tumors, glioblastoma (GBM) is very challenging to treat as chemotherapeutic drugs can only penetrate the brain to a limited extent due to the blood-brain barrier (BBB). Nanoparticles can be an attractive solution for the treatment of GBM as they can transport drugs across the BBB into the tumor. In this study, normal and GBM organoids comprising six brain cell types were developed and applied to study the uptake, BBB penetration, distribution, and efficacy of fluorescent, ultrasmall gold nanoparticles (AuTio-Dox-AF647s) conjugated with doxorubicin (Dox) and AlexaFluor-647-cadaverine (AF647) by confocal laser scanning microscopy (CLSM), using a mixture of dissolved doxorubicin and fluorescent AF647 molecules as a control. It was shown that the nanoparticles could easily penetrate the BBB and were found in normal and GBM organoids, while the dissolved Dox and AF647 molecules alone were unable to penetrate the BBB. Flow cytometry showed a reduction in glioblastoma cells after treatment with AuTio-Dox nanoparticles, as well as a higher uptake of these nanoparticles by GBM cells in the GBM model compared to astrocytes in the normal cell organoids. In summary, our results show that ultrasmall gold nanoparticles can serve as suitable carriers for the delivery of drugs into organoids to study BBB function.
Collapse
Affiliation(s)
- Kathrin Kostka
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45141 Essen, Germany
| | - Viktoriya Sokolova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45141 Essen, Germany
| | - Aya El-Taibany
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Benedikt Kruse
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45141 Essen, Germany
| | - Daniel Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Natalie Wolff
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45141 Essen, Germany
| | - Oleg Prymak
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45141 Essen, Germany
| | - Michael C. Seeds
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45141 Essen, Germany
| | - Anthony J. Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
44
|
Chen X, Zheng Y, Zhang Q, Chen Q, Chen Z, Wu D. Dual-targeted delivery of temozolomide by multi-responsive nanoplatform via tumor microenvironment modulation for overcoming drug resistance to treat glioblastoma. J Nanobiotechnology 2024; 22:264. [PMID: 38760771 PMCID: PMC11100207 DOI: 10.1186/s12951-024-02531-3] [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: 01/14/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor with low survival rate. Currently, temozolomide (TMZ) is the first-line drug for GBM treatment of which efficacy is unfortunately hindered by short circulation time and drug resistance associated to hypoxia and redox tumor microenvironment. Herein, a dual-targeted and multi-responsive nanoplatform is developed by loading TMZ in hollow manganese dioxide nanoparticles functionalized by polydopamine and targeting ligands RAP12 for photothermal and receptor-mediated dual-targeted delivery, respectively. After accumulated in GBM tumor site, the nanoplatform could respond to tumor microenvironment and simultaneously release manganese ion (Mn2+), oxygen (O2) and TMZ. The hypoxia alleviation via O2 production, the redox balance disruption via glutathione consumption and the reactive oxygen species generation, together would down-regulate the expression of O6-methylguanine-DNA methyltransferase under TMZ medication, which is considered as the key to drug resistance. These strategies could synergistically alleviate hypoxia microenvironment and overcome TMZ resistance, further enhancing the anti-tumor effect of chemotherapy/chemodynamic therapy against GBM. Additionally, the released Mn2+ could also be utilized as a magnetic resonance imaging contrast agent for monitoring treatment efficiency. Our study demonstrated that this nanoplatform provides an alternative approach to the challenges including low delivery efficiency and drug resistance of chemotherapeutics, which eventually appears to be a potential avenue in GBM treatment.
Collapse
Affiliation(s)
- Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The First Affiliated Hospital, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The First Affiliated Hospital, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qi Zhang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The First Affiliated Hospital, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qi Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The First Affiliated Hospital, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The First Affiliated Hospital, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, The First Affiliated Hospital, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
45
|
Zhou J, Zheng H, Zhang H, Yu W, Li B, Ye L, Wang L. MCM5 is a Novel Therapeutic Target for Glioblastoma. Onco Targets Ther 2024; 17:371-381. [PMID: 38765057 PMCID: PMC11100520 DOI: 10.2147/ott.s457600] [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: 03/18/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
Objective MCM5 is a DNA licensing factor involved in cell proliferation and has been previously established as an excellent biomarker in a number of malignancies. Nevertheless, the role of MCM5 in GBM has not been fully clarified. The present study aimed to investigate the potential roles of MCM5 in the treatment of GBM and to elucidate its underlying mechanism, which is beneficial for developing new therapeutic strategies and predicting prognosis. Methods Firstly, we obtained transcriptomic and proteomic data from the TCGA and CPTAC databases on glioma patients. Employing the DeSeq2 R package, we then identified genes with joint differential expression in GBM tissues subjected to chemotherapy. To develop a prognostic risk score model, we performed univariate and multivariate Cox regression analyses. In vitro knockdown and overexpression of MCM5 were used to further investigate the biological functions of GBM cells. Additionally, we also delved into the upstream regulation of MCM5, revealing associations with several transcription factors. Finally, we investigated differences in immune cell infiltration and drug sensitivity across diverse risk groups identified in the prognostic risk model. Results In this study, the chemotherapy-treated GBM samples exhibited consistent alterations in 46 upregulated and 94 downregulated genes at both the mRNA and protein levels. Notably, MCM5 emerged as a gene with prognostic significance as well as potential therapeutic relevance. In vitro experiments subsequently validated the role of increased MCM5 expression in promoting GBM cell proliferation and resistance to TMZ. Correlations with transcription factors such as CREB1, CTCF, NFYB, NRF1, PBX1, TEAD1, and USF1 were discovered during upstream regulatory analysis, enriching our understanding of MCM5 regulatory mechanisms. The study additionally delves into immune cell infiltration and drug sensitivity, providing valuable insights for personalized treatment approaches. Conclusion This study identifies MCM5 as a key player in GBM, demonstrating its prognostic significance and potential therapeutic relevance by elucidating its role in promoting cell proliferation and resistance to chemotherapy.
Collapse
Affiliation(s)
- Jian Zhou
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, People’s Republic of China
| | - Housheng Zheng
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Huiru Zhang
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Wenqiang Yu
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Baoer Li
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Liang Ye
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Lu Wang
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| |
Collapse
|
46
|
Liu X, Wu L, Wang L, Li Y. Identification and classification of glioma subtypes based on RNA-binding proteins. Comput Biol Med 2024; 174:108404. [PMID: 38582000 DOI: 10.1016/j.compbiomed.2024.108404] [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: 12/04/2023] [Revised: 03/23/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Glioma is a common and aggressive primary malignant cancer known for its high morbidity, mortality, and recurrence rates. Despite this, treatment options for glioma are currently restricted. The dysregulation of RBPs has been linked to the advancement of several types of cancer, but their precise role in glioma evolution is still not fully understood. This study sought to investigate how RBPs may impact the development and prognosis of glioma, with potential implications for prognosis and therapy. METHODS RNA-seq profiles of glioma and corresponding clinical data from the CGGA database were initially collected for analysis. Unsupervised clustering was utilized to identify crucial tumor subtypes in glioma development. Subsequent time-series analysis and MS model were employed to track the progression of these identified subtypes. RBPs playing a significant role in glioma progression were then pinpointed using WGCNA and Lasso Cox regression models. Functional analysis of these key RBP-related genes was conducted through GSEA. Additionally, the CIBERSORT algorithm was utilized to estimate immune infiltrating cells, while the STRING database was consulted to uncover potential mechanisms of the identified biomarkers. RESULTS Six tumor subgroups were identified and found to be highly homogeneous within each subgroup. The progression stages of these tumor subgroups were determined using time-series analysis and a MS model. Through WGCNA, Lasso Cox, and multivariate Cox regression analysis, it was confirmed that BCLAF1 is correlated with survival in glioma patients and is closely linked to glioma progression. Functional annotation suggests that BCLAF1 may impact glioma progression by influencing RNA splicing, which in turn affects the cell cycle, Wnt signaling pathway, and other cancer development pathways. CONCLUSIONS The study initially identified six subtypes of glioma progression and assessed their malignancy ranking. Furthermore, it was determined that BCLAF1 could serve as an RBP-related prognostic marker, offering significant implications for the clinical diagnosis and personalized treatment of glioma.
Collapse
Affiliation(s)
- Xudong Liu
- School of Medicine, Chongqing University, Chongqing, 400044, China; Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Lei Wang
- College of Life Sciences, Xinyang Normal University, Xinyang, 464000, China.
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
47
|
Horváth L, Biri-Kovács B, Baranyai Z, Stipsicz B, Méhes E, Jezsó B, Krátký M, Vinšová J, Bősze S. New Salicylanilide Derivatives and Their Peptide Conjugates as Anticancer Compounds: Synthesis, Characterization, and In Vitro Effect on Glioblastoma. ACS OMEGA 2024; 9:16927-16948. [PMID: 38645331 PMCID: PMC11024950 DOI: 10.1021/acsomega.3c05727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 04/23/2024]
Abstract
Pharmacologically active salicylanilides (2-hydroxy-N-phenylbenzamides) have been a promising area of interest in medicinal chemistry-related research for quite some time. This group of compounds has shown a wide spectrum of biological activities, including but not limited to anticancer effects. In this study, substituted salicylanilides were chosen to evaluate the in vitro activity on U87 human glioblastoma (GBM) cells. The parent salicylanilide, salicylanilide 5-chloropyrazinoates, a 4-aminosalicylic acid derivative, and the new salicylanilide 4-formylbenzoates were chemically and in vitro characterized. To enhance the internalization of the compounds, they were conjugated to delivery peptides with the formation of oxime bonds. Oligotuftsins ([TKPKG]n, n = 1-4), the ligands of neuropilin receptors, were used as GBM-targeting carrier peptides. The in vitro cellular uptake, intracellular localization, and penetration ability on tissue-mimicking models of the fluorescent peptide derivatives were determined. The compounds and their peptide conjugates significantly decreased the viability of U87 glioma cells. Salicylanilide compound-induced GBM cell death was associated with activation of autophagy, as characterized by immunodetection of autophagy-related processing of light chain 3 protein.
Collapse
Affiliation(s)
- Lilla Horváth
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Beáta Biri-Kovács
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Zsuzsa Baranyai
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Bence Stipsicz
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
- Institute
of Biology, Doctoral School of Biology, Eötvös Loránd University, Budapest 1117, Hungary
| | - Előd Méhes
- Institute
of Physics, Department of Biological Physics, Eötvös Loránd University, Budapest 1117, Hungary
| | - Bálint Jezsó
- Research
Centre for Natural Sciences, Institute of
Enzymology, Budapest 1053, Hungary
- ELTE-MTA
“Momentum” Motor Enzymology Research Group, Department
of Biochemistry, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Martin Krátký
- Department
of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec
Králové, Charles University, 500 03 Hradec Králové, Czech Republic
| | - Jarmila Vinšová
- Department
of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec
Králové, Charles University, 500 03 Hradec Králové, Czech Republic
| | - Szilvia Bősze
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| |
Collapse
|
48
|
Marrocco F, Falvo E, Mosca L, Tisci G, Arcovito A, Reccagni A, Limatola C, Bernardini R, Ceci P, D'Alessandro G, Colotti G. Nose-to-brain selective drug delivery to glioma via ferritin-based nanovectors reduces tumor growth and improves survival rate. Cell Death Dis 2024; 15:262. [PMID: 38615026 PMCID: PMC11016100 DOI: 10.1038/s41419-024-06653-2] [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: 10/04/2023] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 04/15/2024]
Abstract
Gliomas are among the most fatal tumors, and the available therapeutic options are very limited. Additionally, the blood-brain barrier (BBB) prevents most drugs from entering the brain. We designed and produced a ferritin-based stimuli-sensitive nanocarrier with high biocompatibility and water solubility. It can incorporate high amounts of the potent topoisomerase 1 inhibitor Genz-644282. Here, we show that this nanocarrier, named The-0504, can cross the BBB and specifically deliver the payload to gliomas that express high amounts of the ferritin/transferrin receptor TfR1 (CD71). Intranasal or intravenous administration of The-0504 both reduce tumor growth and improve the survival rate of glioma-bearing mice. However, nose-to-brain administration is a simpler and less invasive route that may spare most of the healthy tissues compared to intravenous injections. For this reason, the data reported here could pave the way towards a new, safe, and direct ferritin-based drug delivery method for brain diseases, especially brain tumors.
Collapse
Affiliation(s)
- Francesco Marrocco
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Elisabetta Falvo
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy
| | - Luciana Mosca
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | - Giada Tisci
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Alice Reccagni
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute 17 Pasteur Italia, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
| | - Roberta Bernardini
- Dipartimento di Scienze Cliniche e Medicina Traslazionale Università degli Studi di Roma "Tor Vergata", Rome, Italy
| | - Pierpaolo Ceci
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
- Thena Biotech, Latina, Italy.
| | - Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.
- IRCCS Neuromed, Pozzilli, IS, Italy.
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
| |
Collapse
|
49
|
Rinaldi A, Dumas F, Duskey JT, Imbriano C, Belluti S, Roy C, Ottonelli I, Vandelli MA, Ruozi B, Garcion E, Tosi G, Boury F. Polymer-lipid hybrid nanomedicines to deliver siRNA in and against glioblastoma cells. Int J Pharm 2024; 654:123994. [PMID: 38484859 DOI: 10.1016/j.ijpharm.2024.123994] [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: 12/01/2023] [Revised: 02/27/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Small interfering RNA (siRNA) holds great potential to treat many difficult-to-treat diseases, but its delivery remains the central challenge. This study aimed at investigating the suitability of polymer-lipid hybrid nanomedicines (HNMeds) as novel siRNA delivery platforms for locoregional therapy of glioblastoma. Two HNMed formulations were developed from poly(lactic-co-glycolic acid) polymer and a cationic lipid: 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) or 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Chol). After characterization of the HNMeds, a model siRNA was complexed onto their surface to form HNMed/siRNA complexes. The physicochemical properties and siRNA binding ability of complexes were assessed over a range of nitrogen-to-phosphate (N/P) ratios to optimize the formulations. At the optimal N/P ratio of 10, complexes effectively bound siRNA and improved its protection from enzymatic degradation. Using the NIH3T3 mouse fibroblast cell line, DOTAP-based HNMeds were shown to possess higher cytocompatibility in vitro over the DC-Chol-based ones. As proof-of-concept, uptake and bioefficacy of formulations were also assessed in vitro on U87MG human glioblastoma cell line expressing luciferase gene. Complexes were able to deliver anti-luciferase siRNA and induce a remarkable suppression of gene expression. Noteworthy, the effect of DOTAP-based formulation was not only about three-times higher than DC-Chol-based one, but also comparable to lipofectamine model transfection reagent. These findings set the basis to exploit this nanosystem for silencing relevant GB-related genes in further in vitro and in vivo studies.
Collapse
Affiliation(s)
- Arianna Rinaldi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy; Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France
| | - Florence Dumas
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 213/D, 41125 Modena, Italy
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 213/D, 41125 Modena, Italy
| | - Charlotte Roy
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France
| | - Ilaria Ottonelli
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Maria Angela Vandelli
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Barbara Ruozi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Emmanuel Garcion
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Frank Boury
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France.
| |
Collapse
|
50
|
Esteves L, Caramelo F, Roda D, Carreira IM, Melo JB, Ribeiro IP. Identification of Novel Molecular and Clinical Biomarkers of Survival in Glioblastoma Multiforme Patients: A Study Based on The Cancer Genome Atlas Data. BIOMED RESEARCH INTERNATIONAL 2024; 2024:5582424. [PMID: 38606198 PMCID: PMC11008977 DOI: 10.1155/2024/5582424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/14/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent type of brain tumour; although advancements in treatment have been made, the median survival time for GBM patients has persisted at 15 months. This study is aimed at investigating the genetic alterations and clinical features of GBM patients to find predictors of survival. GBM patients' methylation and gene expression data along with clinical information from TCGA were retrieved. The most overrepresented pathways were identified independently for each omics dataset. From the genes found in at least 30% of these pathways, one gene that was identified in both sets was further examined using the Kaplan-Meier method for survival analysis. Additionally, three groups of patients who started radio and chemotherapy at different times were identified, and the influence of these variations in treatment modality on patient survival was evaluated. Four pathways that seemed to negatively impact survival and two with the opposite effect were identified. The methylation status of PRKCB was highlighted as a potential novel biomarker for patient survival. The study also found that treatment with chemotherapy prior to radiotherapy can have a significant impact on patient survival, which could lead to improvements in clinical management and therapeutic approaches for GBM patients.
Collapse
Affiliation(s)
- Luísa Esteves
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Francisco Caramelo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Laboratory of Biostatistics and Medical Informatics, iCBR-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Domingos Roda
- Algarve Radiation Oncology Unit-Joaquim Chaves Saúde (JCS), Faro, Portugal
| | - Isabel Marques Carreira
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Joana Barbosa Melo
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ilda Patrícia Ribeiro
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| |
Collapse
|