1
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
2
|
Li J, Wu W, Ye L, Zheng B. Hyperglycemia as driver of glioblastoma progression: Insights from Mendelian randomization and single-cell transcriptomics. Brain Res 2025; 1858:149636. [PMID: 40210146 DOI: 10.1016/j.brainres.2025.149636] [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: 11/06/2024] [Revised: 03/19/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Hyperglycemia and diabetes may influence GBM progression by altering tumor metabolism and the tumor microenvironment. However, the causal relationship between blood glucose levels and GBM remains unclear. METHODS Mendelian randomization (MR) analysis was performed using GWAS data from the UK Biobank and FinnGen databases, with fasting blood glucose, plasma glucose, cerebrospinal fluid (CSF) glucose, and diabetes as exposures. Single-cell RNA sequencing of GBM mouse models on high-glucose and control diets was conducted to explore the cellular landscape of the tumor microenvironment under hyperglycemic conditions. Additionally, gene set enrichment analysis (GSEA) was performed on transcriptomic data from brain tissues of diabetic patients to assess the activity of GBM-related pathways. RESULTS MR analysis demonstrated a significant genetic relationship between elevated fasting blood glucose and GBM risk, with an odds ratio (OR) of 40.991 (95 % CI: 2.066-813.447, p = 0.015). Type 2 diabetes (T2D) also showed a potential causal link with GBM, with the Weighted Median and Inverse Variance Weighted methods yielding ORs of 2.740 (95 % CI: 1.033-7.273, p = 0.043) and 2.100 (95 % CI: 1.029-4.287, p = 0.042), respectively. Single-cell transcriptomic analysis of GBM mouse models revealed an increased proportion of GBM tumor stem cells and pro-tumorigenic M2 macrophages in the high-glucose diet (HGD) group. GSEA of diabetic patient brain tissue revealed heightened activity of GBM-related pathways, particularly in astrocytes, endothelial cells, and neurons. CONCLUSION These findings suggest that hyperglycemia may actively contribute to GBM progression by promoting cellular changes within the tumor microenvironment and activating GBM-related pathways in brain tissues.
Collapse
Affiliation(s)
- Jin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wenjing Wu
- The Central Laboratory, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Liguo Ye
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
3
|
Bejarano L, Lourenco J, Kauzlaric A, Lamprou E, Costa CF, Galland S, Maas RR, Guerrero Aruffo P, Fournier N, Brouland JP, Hottinger AF, Daniel RT, Hegi ME, Joyce JA. Single-cell atlas of endothelial and mural cells across primary and metastatic brain tumors. Immunity 2025; 58:1015-1032.e6. [PMID: 40107274 DOI: 10.1016/j.immuni.2025.02.022] [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: 06/19/2024] [Revised: 11/06/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Central nervous system (CNS) malignancies include primary tumors, such as gliomas, and brain metastases (BrMs) originating from diverse extracranial cancers. The blood-brain barrier (BBB) is a key structural component of both primary and metastatic brain cancers. Here, we comprehensively analyzed the two major BBB cell types, endothelial and mural cells, across non-tumor brain tissue, isocitrate dehydrogenase (IDH) mutant (IDH mut) low-grade gliomas, IDH wild-type (IDH WT) high-grade glioblastomas (GBMs), and BrMs from various primary tumors. Bulk and single-cell RNA sequencing, integrated with spatial analyses, revealed that GBMs, but not low-grade gliomas, exhibit significant alterations in the tumor vasculature, including the emergence of diverse pathological vascular cell subtypes. However, these alterations are less pronounced in GBMs than in BrMs. Notably, the BrM vasculature shows higher permeability and more extensive interactions with distinct immune cell populations. This vascular atlas presents a resource toward understanding of tumor-specific vascular features in the brain, providing a foundation for developing vascular- and immune-targeting therapies.
Collapse
Affiliation(s)
- Leire Bejarano
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | - Joao Lourenco
- Agora Cancer Research Centre, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Annamaria Kauzlaric
- Agora Cancer Research Centre, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eleni Lamprou
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Catia F Costa
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Sabine Galland
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roeltje R Maas
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Paola Guerrero Aruffo
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Centre, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Andreas F Hottinger
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roy T Daniel
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Monika E Hegi
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|
4
|
Varlamova EG. Roles of selenium-containing glutathione peroxidases and thioredoxin reductases in the regulation of processes associated with glioblastoma progression. Arch Biochem Biophys 2025; 766:110344. [PMID: 39956249 DOI: 10.1016/j.abb.2025.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Glioblastoma remains the most common and aggressive primary tumor of the central nervous system in adults. Current treatment options include standard surgical resection combined with radiation/chemotherapy, but such protocol most likely only delays the inevitable. Therefore, the problem of finding therapeutic targets to prevent the occurrence and development of this severe oncological disease is currently acute. It is known that the functions of selenoproteins in the regulation of carcinogenesis processes are not unambiguous. Either they exhibit cytotoxic activity on cancer cells, or cytoprotective. A special place in the progression of oncological diseases of various etiologies is occupied by proteins of the thioredoxin and glutathione systems. These are two cellular antioxidant systems that regulate redox homeostasis, counteracting the increased production of reactive oxygen species in cells. The review reflects the latest data on the role of key enzymes of these redox systems in the regulation of processes associated with the progression of glioblastoma. A thorough consideration of these issues will expand fundamental knowledge about the functions of selenium-containing thioredoxin reductases and glutathione peroxidases in the therapy of glioblastomas and provide an understanding of the prospects for the treatment of this aggressive oncological disease.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", St. Institutskaya 3, Pushchino, 142290, Russia.
| |
Collapse
|
5
|
Rizwan A, Sridharan B, Park JH, Kim D, Vial JC, Kyhm K, Lim HG. Nanophotonic-enhanced photoacoustic imaging for brain tumor detection. J Nanobiotechnology 2025; 23:170. [PMID: 40045308 PMCID: PMC11881315 DOI: 10.1186/s12951-025-03204-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Photoacoustic brain imaging (PABI) has emerged as a promising biomedical imaging modality, combining high contrast of optical imaging with deep tissue penetration of ultrasound imaging. This review explores the application of photoacoustic imaging in brain tumor imaging, highlighting the synergy between nanomaterials and state of the art optical techniques to achieve high-resolution imaging of deeper brain tissues. PABI leverages the photoacoustic effect, where absorbed light energy causes thermoelastic expansion, generating ultrasound waves that are detected and converted into images. This technique enables precise diagnosis, therapy monitoring, and enhanced clinical screening, specifically in the management of complex diseases such as breast cancer, lymphatic disorder, and neurological conditions. Despite integration of photoacoustic agents and ultrasound radiation, providing a comprehensive overview of current methodologies, major obstacles in brain tumor treatment, and future directions for improving diagnostic and therapeutic outcomes. The review underscores the significance of PABI as a robust research tool and medical method, with the potential to revolutionize brain disease diagnosis and treatment.
Collapse
Affiliation(s)
- Ali Rizwan
- Smart Gym-Based Translational Research Center for Active Senior'S Healthcare, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Badrinathan Sridharan
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Jin Hyeong Park
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Daehun Kim
- Indusrty 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Jean-Claude Vial
- Université Grenoble Alpes, CNRS, LIPhy, 38000, Grenoble, France
- Department of Optics & Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Kwangseuk Kyhm
- Department of Optics & Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Hae Gyun Lim
- Smart Gym-Based Translational Research Center for Active Senior'S Healthcare, Pukyong National University, Busan, 48513, Republic of Korea.
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
- Indusrty 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
6
|
Sun P, Liu F, Huo K, Wang J, Cheng Y, Shang S, Ma W, Yu J, Han J. Adiponectin facilitates the cell cycle, inhibits cell apoptosis and induces temozolomide resistance in glioblastoma via the Akt/mTOR pathway. Oncol Lett 2025; 29:127. [PMID: 39807099 PMCID: PMC11726000 DOI: 10.3892/ol.2025.14875] [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: 07/02/2024] [Accepted: 10/22/2024] [Indexed: 01/16/2025] Open
Abstract
Adiponectin (ADN) regulates DNA synthesis, cell apoptosis and cell cycle to participate in the pathology and progression of glioblastoma. The present study aimed to further explore the effect of ADN on temozolomide (TMZ) resistance in glioblastoma and the underlying mechanism of action. Glioblastoma cell lines (U251 and U87-MG cells) were treated with ADN and TMZ at different concentrations; subsequently, 3.0 µg/ml ADN and 1.0 mM TMZ were selected as the optimal concentrations for the experimental conditions. LY294002 (a PI3K inhibitor) was added to ADN or ADN + TMZ-treated glioblastoma cell lines. Cell growth rate was determined using the Cell Counting Kit-8 assay, the apoptotic rate and cell cycle were evaluated using Annexin V/propidium iodide and cell cycle assays, and p-Akt (Thr308), p-Akt (Ser473), Akt, p-mTOR, c-caspase 3, caspase 3, Bax, cyclin B1 and cyclin D1 expression was determined by western blotting. Adiponectin receptor (ADIPOR) 1 and ADIPOR2 were expressed in glioblastoma cell lines. The glioblastoma cell line growth rate was increased by ADN in a concentration- and time-dependent manner. ADN inhibited glioblastoma cell line apoptosis and facilitated cell cycle. Of note, ADN activated the Akt/mTOR pathway and the addition of LY294002 reversed the effect of ADN, indicating that ADN activated the Akt/mTOR pathway to suppress apoptosis and promote cell cycle in glioblastoma cell lines. Notably, TMZ inhibited glioblastoma cell line growth, promoted apoptosis and increased G2 phase cell cycle arrest. However, the addition of ADN reversed the effect of TMZ in glioblastoma cell lines, disclosing that ADN induced TMZ resistance. Markedly, ADN-mediated TMZ resistance was further attenuated by LY294002, suggesting that ADN activated the Akt/mTOR pathway to induce TMZ resistance in glioblastoma cell lines. In conclusion, ADN activated the Akt/mTOR pathway to facilitate cell cycle, inhibit cell apoptosis and induce TMZ resistance in glioblastoma.
Collapse
Affiliation(s)
- Peng Sun
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fude Liu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianyi Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yawen Cheng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenlong Ma
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jia Yu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianfeng Han
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
7
|
Tang ZQ, Ye YR, Shen Y. Molecular Mechanisms and Strategies for Inducing Neuronal Differentiation in Glioblastoma Cells. Cell Reprogram 2025; 27:24-32. [PMID: 39880036 DOI: 10.1089/cell.2024.0087] [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: 01/31/2025] Open
Abstract
Glioblastoma multiforme (GBM) is a highly invasive brain tumor, and traditional treatments combining surgery with radiochemotherapy have limited effects, with tumor recurrence being almost inevitable. Given the lack of proliferative capacity in neurons, inducing terminal differentiation of GBM cells or glioma stem cells (GSCs) into neuron-like cells has emerged as a promising strategy. This approach aims to suppress their proliferation and self-renewal capabilities through differentiation. This review summarizes the methods involved in recent research on the neuronal differentiation of GBM cells or GSCs, including the regulation of transcription factors, signaling pathways, miRNA, and the use of small molecule drugs, among various strategies. It also outlines the interconnections between the mechanisms studied, hoping to provide ideas for exploring new therapeutic avenues for GBM and the development of differentiation-inducing drugs for GBM.
Collapse
Affiliation(s)
- Zhao-Qi Tang
- Department of Pharmacy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, China
| | - Yan-Rong Ye
- Department of Pharmacy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shen
- Department of Pharmacy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Rusak A, Wiatrak B, Krawczyńska K, Górnicki T, Zagórski K, Zadka Ł, Fortuna W. Starting points for the development of new targeted therapies for glioblastoma multiforme. Transl Oncol 2025; 51:102187. [PMID: 39531784 PMCID: PMC11585793 DOI: 10.1016/j.tranon.2024.102187] [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: 06/03/2024] [Revised: 09/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive and lethal brain tumors, characterized by rapid growth, invasiveness, and resistance to standard therapies, including surgery, chemotherapy, and radiotherapy. Despite advances in treatment, GBM remains highly resistant due to its complex molecular mechanisms, including angiogenesis, invasion, immune modulation, and lipid metabolism dysregulation. This review explores recent breakthroughs in targeted therapies, focusing on innovative drug carriers such as nanoparticles and liposomes, and their potential to overcome GBM's chemo- and radioresistant phenotypes. We also discuss the molecular pathways involved in GBM progression and the latest therapeutic strategies, including immunotherapy and precision medicine approaches, which hold promise for improving clinical outcomes. The review highlights the importance of understanding GBM's genetic and molecular heterogeneity to develop more effective, personalized treatment protocols aimed at increasing survival rates and enhancing the quality of life for GBM patients.
Collapse
Affiliation(s)
- Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland.
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, J. Mikulicza-Radeckiego 2 Street, Wroclaw 50-345, Poland.
| | - Klaudia Krawczyńska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland.
| | - Tomasz Górnicki
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland
| | - Karol Zagórski
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland
| | - Łukasz Zadka
- Division of Ultrastructural Research, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland; Department of Clinical Pharmacology, Wroclaw Medical University, Borowska 211a, Wroclaw 50-556, Poland.
| | - Wojciech Fortuna
- Department of Neurosurgery, Wroclaw Medical University, Borowska 213St, Wroclaw 50-556, Poland.
| |
Collapse
|
9
|
Bostancı F, Şengelen A, Aksüt Y, Yıldırım E, Öğütcü İ, Yücel O, Emik S, Gürdağ G, Pekmez M. Indomethacin-encapsulated PLGA nanoparticles improve therapeutic efficacy by increasing apoptosis and reducing motility in glioblastoma cells. Pharm Dev Technol 2025; 30:25-36. [PMID: 39750021 DOI: 10.1080/10837450.2024.2448333] [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: 09/25/2024] [Revised: 12/02/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Glioblastoma, with a low survival rate, is an aggressive and difficult-to-treat lethal type of brain cancer. Indomethacin (IND), a non-steroidal anti-inflammatory drug, has antitumoral activity in many cancers, including gliomas. However, its poor aqueous solubility is a critical issue. Nanomaterials are crucial tools for overcoming solubility problems and facilitating drug delivery. Herein, a polymeric nanoparticle system, poly(lactic-co-glycolic acid) (PLGA) was used to encapsulate IND. Although PLGA is an FDA-approved copolymer for drug delivery, no trials with IND-loaded PLGA-NPs have been conducted to treat brain tumors. Encapsulation success was revealed by DLS, zeta potential, TEM, and FTIR analysis; IND/PLGA-NPs had nanoscale particle size (160.6 nm), narrow size distribution (0.230, PDI), and good stability (-23.9 mV). Fluorescence imaging showed that PLGA-NPs can penetrate U-87MG cells. Short-term/one-hour treatment with bound-IND increased the free-IND effect in gliomas by ⁓10 times/48h and 12.39 times/72h. Even against long-term exposure to IND, IND/PLGA-NP treatment revealed a highly marked result; the IC50 value of bound-IND (treatment-time:1h, analysis at 48h) was ∼200µM, IC50 value of free-IND (treatment-time:48h) was ∼390µM. Furthermore, IND/PLGA-NPs' anticancer activity (100 µM of IND/1h, analysis at 48h) was also supported by induced apoptosis and reduced migration/colony formation in glioma cells. All evidence suggests that IND/PLGA-NPs may be a potentially promising agent for treating gliomas.
Collapse
Affiliation(s)
- Ferhat Bostancı
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkiye
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Istanbul, Turkiye
| | - Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye
| | - Yunus Aksüt
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye
- Department of Molecular Biology and Genetics, Basic Medical Sciences, School of Medicine, Koç University, Istanbul, Turkiye
| | - Eren Yıldırım
- Department of Chemical Engineering, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
| | - İrem Öğütcü
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkiye
| | - Oğuz Yücel
- Department of Chemical Engineering, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
| | - Serkan Emik
- Department of Chemical Engineering, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
| | - Gülten Gürdağ
- Department of Chemical Engineering, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
| | - Murat Pekmez
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye
| |
Collapse
|
10
|
Min Z, Guo Y, Ning L. Paromomycin targets HDAC1-mediated SUMOylation and IGF1R translocation in glioblastoma. Front Pharmacol 2024; 15:1490878. [PMID: 39723246 PMCID: PMC11668589 DOI: 10.3389/fphar.2024.1490878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/08/2024] [Indexed: 12/28/2024] Open
Abstract
Objective This study investigates the effects of Paromomycin on SUMOylation-related pathways in glioblastoma (GBM), specifically targeting HDAC1 inhibition. Methods Using TCGA and GTEx datasets, we identified SUMOylation-related genes associated with GBM prognosis. Molecular docking analysis suggested Paromomycin as a potential HDAC1 inhibitor. In vitro assays on U-251MG GBM cells were performed to assess Paromomycin's effects on cell viability, SUMOylation gene expression, and IGF1R translocation using CCK8 assays, qRT-PCR, and immunofluorescence. Results Paromomycin treatment led to a dose-dependent reduction in GBM cell viability, colony formation, and migration. It modulated SUMO1 expression and decreased IGF1R nuclear translocation, an effect reversible by the HDAC1 inhibitor Trochostatin A (TSA), suggesting Paromomycin's involvement in SUMO1-regulated pathways. Conclusion This study highlights Paromomycin's potential as a therapeutic agent for GBM by targeting HDAC1-mediated SUMOylation pathways and influencing IGF1R translocation, warranting further investigation for its clinical application.
Collapse
|
11
|
Şengelen A, Önay-Uçar E. Rosmarinic acid attenuates glioblastoma cells and spheroids' growth and EMT/stem-like state by PTEN/PI3K/AKT downregulation and ERK-induced apoptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156060. [PMID: 39341126 DOI: 10.1016/j.phymed.2024.156060] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Glioblastoma (GB) is a highly malignant type of brain cancer with a poor prognosis. Therapeutic strategies for GB are still limited. Rosmarinic acid (RA), a polyphenolic compound, is a promising experimental anticancer agent, but its specific protein targets for GB remain unclear. PURPOSE This study aimed to elucidate the anticancer effects of RA in 2D- and 3D-GB cells and the underlying mechanisms. METHODS 3D-tumor spheroids (mimics in vivo tumors) were obtained by the hanging-drop/agarose method. RA's anti-glioma activity on U-87MG (p53-wt/PTEN-mt) and LN229 (p53-mt/PTEN-wt) cells was evaluated through cell viability, colony-formation, migration/invasion/angiogenesis assays, fluorescence imaging, and spheroid growth analysis. The underlying mechanism of the anticancer effects of RA was investigated by Western blot and immunofluorescence analysis. The MEK inhibitor U0126 was used to block ERK phosphorylation. RESULTS RA treatments exerted anti-proliferative and pro-apoptotic effects on human GB cells. RA dose-dependently reduced angiogenesis and intracellular ROS levels, suppressed glioma growth, and migration/invasion in 2D-culture and cancer stem cell (CSC)-like 3D-spheroid culture (SPC). Repeated therapy in SPC was more effective by leading to disrupted structure than a single treatment. Treatments in SPC also suppressed epithelial-mesenchymal transition (EMT) and CSC-like properties. Strikingly, RA downregulated the SIRT1/FOXO1/NF-κB axis independently of p53 or PTEN function in both gliomas. Immunofluorescence labeling revealed decreased SIRT1 and NF-κB-p65 and increased FOXO1 and GAPDH proteins in nuclear location (associated with apoptosis). Surprisingly, RA increased p-ERK1/2 levels, but priming with U0126 abolished RA-mediated p-ERK upregulation; thus, autophagy and apoptosis induction in GB cells were prevented, and the growth of GB spheroids accelerated. Specifically, RA also inhibited the PTEN/PI3K/AKT pathway in U-87MG cells. Due to genetic differences in cells, U-87MG cells were more sensitive to RA treatments than LN229 cells. Meanwhile, our positive control drug trial results with FDA-approved temozolomide (TMZ) used in GB treatment showed that our test compound rosmarinic acid exhibited higher therapeutic effects than TMZ at lower doses. CONCLUSION Suppression of EMT, downregulation of SIRT1/FOXO1/NF-κB axis, inhibition of PTEN/PI3K/AKT signaling pathway, and ERK-induced apoptosis and autophagy were determined to be involved in stopping glioma progression. Our findings for the first time, revealed that RA may have potential therapeutic use by having multiple targets in human brain cancer with further clinical studies.
Collapse
Affiliation(s)
- Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkiye.
| | - Evren Önay-Uçar
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye
| |
Collapse
|
12
|
Massarotti M, Corna P, Mallik A, Milanesi G, Casali C, Magrassi L, Comincini S. Development and Biological Characterization of Cancer Biomimetic Membrane Nanovesicles for Enhancing Therapy Efficacy in Human Glioblastoma Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1779. [PMID: 39591021 PMCID: PMC11597144 DOI: 10.3390/nano14221779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024]
Abstract
As nanocarriers of a new generation, biomimetic nanovesicles are an emerging class of therapeutic tools whose surface is integrated or fabricated with biomaterials capable of mimicking the biological features and functions of native cells. Thanks to this, biomimetic nanovesicles, in particular, those made by plasma membrane moieties, possess greatly improved biocompatibility, high target specificity, a long retention time, and minimal undesired immune responses. For these reasons, a multitude of progenitor cells including cancer ones were employed as templates to generate biomimetic or membrane-camouflaged nanovesicles hosting different therapeutic compounds. In this contribution, different membrane-derived biomimetic vesicles (M-NVs) were generated by osmotic lysis or plasma membrane isolation approaches from normal and cancer cell lines and assayed against in vitro models of human glioblastoma. M-NVs were compared in their cellular internalization degrees of DNA and proteins, morphologically and molecularly characterized, expressing an extracellular membrane-associated marker. Then, Rose Bengal (RB), a photoactivable drug characterized by a relatively low cellular uptake, was incorporated into nascent glioblastoma-derived M-NVs and finally administered to homotypic receiving cells, showing an increased degree of internalization as well as induced cytotoxic effects, even in the absence of photodynamic direct stimulation. Similar results were also obtained assaying lyophilized M-NVs loaded with RB. In conclusion, M-NVs generated by cell membranes effectively deliver several cargoes, including therapeutic molecules, maintain functionality after lyophilization, and show significant internalization effects, making them a promising strategy for therapeutic applications against human glioblastoma cells.
Collapse
Affiliation(s)
- Martina Massarotti
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (M.M.); (A.M.); (G.M.); (C.C.)
| | - Paola Corna
- Department of Clinical Surgical Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (P.C.); (L.M.)
| | - Aromita Mallik
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (M.M.); (A.M.); (G.M.); (C.C.)
| | - Gloria Milanesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (M.M.); (A.M.); (G.M.); (C.C.)
| | - Claudio Casali
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (M.M.); (A.M.); (G.M.); (C.C.)
| | - Lorenzo Magrassi
- Department of Clinical Surgical Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (P.C.); (L.M.)
| | - Sergio Comincini
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (M.M.); (A.M.); (G.M.); (C.C.)
| |
Collapse
|
13
|
Yuan F, Wang Y, Yuan L, Ye L, Hu Y, Cheng H, Li Y. Machine learning-based new classification for immune infiltration of gliomas. PLoS One 2024; 19:e0312071. [PMID: 39453922 PMCID: PMC11508054 DOI: 10.1371/journal.pone.0312071] [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: 06/03/2024] [Accepted: 09/30/2024] [Indexed: 10/27/2024] Open
Abstract
BACKGROUND Glioma is a highly heterogeneous and poorly immunogenic malignant tumor, with limited efficacy of immunotherapy. The characteristics of the immunosuppressive tumor microenvironment (TME) are one of the important factors hindering the effectiveness of immunotherapy. Therefore, this study aims to reveal the immune microenvironment (IME) characteristics of glioma and predict different immune subtypes using machine learning methods, providing guidance for immune therapy in glioma. METHODS We first performed unsupervised cluster analysis on the genes and arrays of 693 gliomas in CGGA database and 702 gliomas in TCGA database. Then establish and verify the classification model through Machine Learning (ML). Then, use DAVID to perform functional enrichment analysis for different immune subtypes. Next step, analyze the immune cell distribution, stemness maintenance, mesenchymal phenotype, neuronal phenotype, tumorigenic cytokines, molecular and clinical characteristics of different immune subtypes of gliomas. RESULTS Firstly, we divide the IME of gliomas in the CGGA database into four different subtypes, namely IM1, IM2, IM3, and IM4; similarly, the IME of gliomas in the TCGA database can also be divided into four different subtypes (IMA, IMB, IMC, and IMD). Next, based on ML, we developed a highly reliable model for predicting different immune subtypes of glioma. Then, we found that Monocytic lineage, Myeloid dendritic cells, NK cells and CD8 T cells had the highest enrichment in the IM1/IMD subtypes. Cytotoxic lymphocytes were highest expressed in the IM4/IMA subtypes. Next step, Enrichment analysis revealed that the IM1-IMD subtypes were mainly closely related to the production and secretion of IL-8 and TNF signaling pathway. The IM2-IMB subtypes were strongly associated with leukocyte activation and NK cell mediated cytotoxicity. The IM3-IMC subtypes were closely related to mitotic nuclear division and mitotic cell cycle process. The IM4-IMA subtypes were strongly associated with Central Nervous System (CNS) development and striated muscle tissue development. Afterwards, Single sample gene set enrichment analysis (ssGSEA) showed that stemness maintenance phenotypes were mainly enriched in the IM4/IMA subtypes; Neuronal phenotypes were closely associated with the IM2/IMB subtypes; and mesenchymal phenotypes and tumorigenic cytokines were highly correlated with the IM2 /IMB subtypes. Finally, we found that compared with patients in the IM2/IMB and IM4/IMA subtypes, the IM1/IMD and IM3/IMC subtypes have the highest proportion of GBM patients, the shortest average overall survival of patients and the lowest proportion of patients with IDH mutation and 1p36/19q13 co-deletion. CONCLUSIONS We developed a highly reliable model for predicting different immune subtypes of glioma by ML. Then, we comprehensively analyzed the immune infiltration, molecular and clinical features of different immune subtypes of gliomas and defined gliomas into four subtypes: immunogenic subtype, adaptive immune resistance subtype, mesenchymal subtype, and immune tolerance subtype, which represent different TMEs and different stages of tumor development.
Collapse
Affiliation(s)
- Feng Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yingshuai Wang
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians- University Munich, Munich, Germany
| | - Lei Yuan
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Ye
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yangchun Hu
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hongwei Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Li
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
14
|
Noor L, Upadhyay A, Joshi V. Role of T Lymphocytes in Glioma Immune Microenvironment: Two Sides of a Coin. BIOLOGY 2024; 13:846. [PMID: 39452154 PMCID: PMC11505600 DOI: 10.3390/biology13100846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Glioma is known for its immunosuppressive microenvironment, which makes it challenging to target through immunotherapies. Immune cells like macrophages, microglia, myeloid-derived suppressor cells, and T lymphocytes are known to infiltrate the glioma tumor microenvironment and regulate immune response distinctively. Among the variety of immune cells, T lymphocytes have highly complex and multifaceted roles in the glioma immune landscape. T lymphocytes, which include CD4+ helper and CD8+ cytotoxic T cells, are known for their pivotal roles in anti-tumor responses. However, these cells may behave differently in the highly dynamic glioma microenvironment, for example, via an immune invasion mechanism enforced by tumor cells. Therefore, T lymphocytes play dual roles in glioma immunity, firstly by their anti-tumor responses, and secondly by exploiting gliomas to promote immune invasion. As an immunosuppression strategy, glioma induces T-cell exhaustion and suppression of effector T cells by regulatory T cells (Tregs) or by altering their signaling pathways. Further, the expression of immune checkpoint inhibitors on the glioma cell surface leads to T cell anergy and dysfunction. Overall, this dynamic interplay between T lymphocytes and glioma is crucial for designing more effective immunotherapies. The current review provides detailed knowledge on the roles of T lymphocytes in the glioma immune microenvironment and helps to explore novel therapeutic approaches to reinvigorate T lymphocytes.
Collapse
Affiliation(s)
- Laiba Noor
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Vibhuti Joshi
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| |
Collapse
|
15
|
Hasan S, Mahmud Z, Hossain M, Islam S. Harnessing the role of aberrant cell signaling pathways in glioblastoma multiforme: a prospect towards the targeted therapy. Mol Biol Rep 2024; 51:1069. [PMID: 39424705 DOI: 10.1007/s11033-024-09996-3] [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: 06/19/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Glioblastoma Multiforme (GBM), designated as grade IV by the World Health Organization, is the most aggressive and challenging brain tumor within the central nervous system. Around 80% of GBM patients have a poor prognosis, with a median survival of 12-15 months. Approximately 90% of GBM cases originate from normal glial cells via oncogenic processes, while the remainder arise from low-grade tumors. GBM is notorious for its heterogeneity, high recurrence rates, invasiveness, and aggressive behavior. Its malignancy is driven by increased invasive migration, proliferation, angiogenesis, and reduced apoptosis. Throughout various stages of central nervous system (CNS) development, pivotal signaling pathways, including Wnt/β-catenin, Sonic hedgehog signaling (Shh), PI3K/AKT/mTOR, Ras/Raf/MAPK/ERK, STAT3, NF-КB, TGF-β, and Notch signaling, orchestrate the growth, proliferation, differentiation, and migration of neural progenitor cells in the brain. Numerous upstream and downstream regulators within these signaling pathways have been identified as significant contributors to the development of human malignancies. Disruptions or aberrant activations in these pathways are linked to gliomagenesis, enhancing the invasiveness, progression, and aggressiveness of GBM, along with epithelial to mesenchymal transition (EMT) and the presence of glioma stem cells (GSCs). Traditional GBM treatment involves surgery, radiotherapy, and chemotherapy with Temozolomide (TMZ). However, most patients experience tumor recurrence, leading to low survival rates. This review provides an overview of the major cell signaling pathways involved in gliomagenesis. Furthermore, we explore the signaling pathways leading to therapy resistance and target key molecules within these signaling pathways, paving the way for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Subbrina Hasan
- Laboratory of Neuroscience and Neurogenetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Mahmud Hossain
- Laboratory of Neuroscience and Neurogenetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Sohidul Islam
- Department of Biochemistry & Microbiology, North South University, Dhaka, 1229, Bangladesh
| |
Collapse
|
16
|
Wang G, Man Y, Cao K, Zhao L, Lun L, Chen Y, Zhao X, Wang X, Zhang L, Hao C. An immune-related gene pair signature predicts the prognosis and immunotherapeutic response in glioblastoma. Heliyon 2024; 10:e39025. [PMID: 39435104 PMCID: PMC11492119 DOI: 10.1016/j.heliyon.2024.e39025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Background Glioblastoma (GBM) has the feature of aggressive growth and high rates of recurrence. Immunotherapy was not included in standard therapy for GBM due to lacking the predictive biomarkers. In the present study, we performed an immune-related gene pair (IRGP) signature to predict the prognosis and immunotherapy response of GBM. Methods A total of 160 GBM patients from TCGA were included. ssGSEA was conducted to evaluate the immune infiltration level. Univariate Cox, LASSO regression analysis, ROC analysis, and Kaplan-Meier survival analysis were applied to construct and evaluate the risk model. Moreover, the association between immune infiltration and the risk score was assessed. Finally, the expression of immune checkpoints between different risk groups was explored. Results According to the normal/tumor, high-/low-immunity group, we identified 125 differentially expressed immune-related genes. Subsequently, a prognostic model including 22 IRGPs was established. The area under the ROC curve to predict 1, 3, and 5-year was 0.811, 0.958, and 0.99 respectively. According to the optimal cut-off value of the 3-year ROC curve, patients were classified into high- and low-risk groups. The Kaplan-Meier analysis result indicated that patients in the low-risk group have longer survival time. The risk score was an independent prognostic predictor (P < 0.001). Moreover, PDCD1 was positively correlated with the risk score (P < 0.01). We also found that patients with high PDCD1 expression had worse survival. Conclusions The IRGP signature was built to predict the prognosis of GBM patients. This signature can serve as a tool to predict the response to immunotherapy in GBM.
Collapse
Affiliation(s)
- Gang Wang
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingchun Man
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Kui Cao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lihong Zhao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lixin Lun
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yiyang Chen
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xinyu Zhao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueying Wang
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lijie Zhang
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Chuncheng Hao
- Department of Head and Neck Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
17
|
Priya B, Chhabria D, Mahesh Dhongdi J, Kirubakaran S. A novel approach to investigate the combinatorial effects of TLK1 (Tousled-Like Kinase1) inhibitors with Temozolomide for glioblastoma therapy. Bioorg Chem 2024; 151:107643. [PMID: 39029318 DOI: 10.1016/j.bioorg.2024.107643] [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: 05/17/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
Glioblastoma multiforme (GBM) is an aggressive, incurable brain tumor with poor prognosis and limited treatment options. Temozolomide (TMZ) is the standard chemotherapeutic treatment for GBM, but its efficacy has drawn strong criticism from clinicians due to short survival gains and frequent relapses. One critical limitation of TMZ therapy is the hyperactivation of DNA repair pathways, which over time neutralizes the cytotoxic effects of TMZ, thus highlighting the urgent need for new treatment approaches. Addressing this, our study explores the therapeutic potential of in-house-designed phenothiazine-based Tousled-like kinase-1 (TLK1) inhibitors for GBM treatment. TLK1, overexpressed in GBM, plays a role in DNA repair. Phenothiazines are known to cross the blood-brain barrier (BBB). Among all molecules, J54 was identified as a potential lead molecule with improved cytotoxicity. In the context of O6-methylguanine-DNA methyltransferase (MGMT)-deficient GBM cells, the combined administration of phenothiazines and TMZ exhibited a collective reduction in clonogenic growth, coupled with anti-migratory and anti-invasion effects. Conversely, in MGMT-proficient cells, phenothiazine monotherapy alone showed reduced clonogenic growth, along with anti-migratory and anti-invasion effects. Notably, a synergistic increase in γH2AX levels and concurrent attenuation of DNA repair upon combinatorial exposure to TMZ and J54 were observed, implying increased cytotoxicity due to sustained DNA strand breaks. Overall, this study provides new insights into TLK1 inhibition for GBM therapy. Collectively, these findings indicate that TLK1 is one of the upregulated kinases in GBM and phenothiazine-based TLK1 inhibitors could be a promising treatment option for GBM patients.
Collapse
Affiliation(s)
- Bhanu Priya
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj Campus, Gujarat 382355, India
| | - Dimple Chhabria
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj Campus, Gujarat 382355, India
| | - Janhvi Mahesh Dhongdi
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj Campus, Gujarat 382355, India
| | - Sivapriya Kirubakaran
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj Campus, Gujarat 382355, India.
| |
Collapse
|
18
|
Yeon Kim S, Tang M, Lu T, Chih SY, Li W. Ferroptosis in glioma therapy: advancements in sensitizing strategies and the complex tumor-promoting roles. Brain Res 2024; 1840:149045. [PMID: 38821335 PMCID: PMC11323215 DOI: 10.1016/j.brainres.2024.149045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic regulated cell death, is induced by the accumulation of lipid peroxides on cellular membranes. Over the past decade, ferroptosis has emerged as a crucial process implicated in various physiological and pathological systems. Positioned as an alternative modality of cell death, ferroptosis holds promise for eliminating cancer cells that have developed resistance to apoptosis induced by conventional therapeutics. This has led to a growing interest in leveraging ferroptosis for cancer therapy across diverse malignancies. Gliomas are tumors arising from glial or precursor cells, with glioblastoma (GBM) being the most common malignant primary brain tumor that is associated with a dismal prognosis. This review provides a summary of recent advancements in the exploration of ferroptosis-sensitizing methods, with a specific focus on their potential application in enhancing the treatment of gliomas. In addition to summarizing the therapeutic potential, this review also discusses the intricate interplay of ferroptosis and its potential tumor-promoting roles within gliomas. Recognizing these dual roles is essential, as they could potentially complicate the therapeutic benefits of ferroptosis. Exploring strategies aimed at circumventing these tumor-promoting roles could enhance the overall therapeutic efficacy of ferroptosis in the context of glioma treatment.
Collapse
Affiliation(s)
- Soo Yeon Kim
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Tong Lu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Stephen Y Chih
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
19
|
Devi CM, Deka K, Das AK, Talukdar A, Sola P. Recent Advances in Marine-Derived Nanoformulation for the Management of Glioblastoma. Mol Biotechnol 2024:10.1007/s12033-024-01287-3. [PMID: 39327380 DOI: 10.1007/s12033-024-01287-3] [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: 05/17/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
Glioma is the most common and aggressive type of central nervous system tumor as categorized by the World Health Organization. Glioblastoma (GBA), in general, exhibits a grim prognosis and short life expectancy, rarely exceeding 14 months. The dismal prognosis is primarily attributed to the development of chemoresistance to temozolomide, the primary therapeutic agent for GBA treatment. Hence, it becomes imperative to develop novel drugs with antitumor efficacy rooted in distinct mechanisms compared to temozolomide. The vast marine environment contains a wealth of naturally occurring compounds from the sea (known as marine-derived natural products), which hold promise for future research in the quest for new anticancer drugs. Ongoing advancements in anticancer pharmaceuticals have led to an upswing in the isolation and validation of numerous pioneering breakthroughs and improvements in anticancer therapeutics. Nonetheless, the availability of FDA-approved marine-derived anticancer drugs remains limited, owing to various challenges and constraints. Among these challenges, drug delivery is a prominent hurdle. This review delves into an alternative approach for delivering marine-derived drugs using nanotechnological formulations and their mechanism of action for treating GBA.
Collapse
Affiliation(s)
- Chanam Melody Devi
- Department of Pharmaceutics, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Santipur, Kamrup, Assam, 781125, India
| | - Kangkan Deka
- Department of Pharmacognosy, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Santipur, Kamrup, Assam, 781125, India
| | - Amit Kumar Das
- Department of Pharmaceutics, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Santipur, Kamrup, Assam, 781125, India
| | - Apurba Talukdar
- Department of Pharmaceutics, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Santipur, Kamrup, Assam, 781125, India
| | - Piyong Sola
- Department of Pharmacology, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Santipur, Kamrup, Assam, 781125, India.
| |
Collapse
|
20
|
Zhang F, Ye J, Zhu J, Qian W, Wang H, Luo C. Key Cell-in-Cell Related Genes are Identified by Bioinformatics and Experiments in Glioblastoma. Cancer Manag Res 2024; 16:1109-1130. [PMID: 39253064 PMCID: PMC11382672 DOI: 10.2147/cmar.s475513] [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: 04/25/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
Purpose This study aimed to explore the roles of cell-in-cell (CIC)-related genes in glioblastoma (GBM) using bioinformatics and experimental strategies. Patients and Methods The ssGSEA algorithm was used to calculate the CIC score for each patient. Subsequently, differentially expressed genes (DEGs) between the CIClow and CIChigh groups and between the tumor and control samples were screened using the limma R package. Key CIC-related genes (CICRGs) were further filtered using univariate Cox and LASSO analyses, followed by the construction of a CIC-related risk score model. The performance of the risk score model in predicting GBM prognosis was evaluated using ROC curves and an external validation cohort. Moreover, their location and differentiation trajectory in GBM were analyzed at the single-cell level using the Seurat R package. Finally, the expression of key CICRGs in clinical samples was examined by qPCR. Results In the current study, we found that CIC scorelow group had a significantly better survival in the TCGA-GBM cohort, supporting the important role of CICRGs in GBM. Using univariate Cox and LASSO analyses, PTX3, TIMP1, IGFBP2, SNCAIP, LOXL1, SLC47A2, and LGALS3 were identified as key CICRGs. Based on this data, a CIC-related prognostic risk score model was built using the TCGA-GBM cohort and validated in the CGGA-GBM cohort. Further mechanistic analyses showed that the CIC-related risk score is closely related to immune and inflammatory responses. Interestingly, at the single-cell level, key CICRGs were expressed in the neurons and myeloids of tumor tissues and exhibited unique temporal dynamics of expression changes. Finally, the expression of key CICRGs was validated by qPCR using clinical samples from GBM patients. Conclusion We identified novel CIC-related genes and built a reliable prognostic prediction model for GBM, which will provide further basic clues for studying the exact molecular mechanisms of GBM pathogenesis from a CIC perspective.
Collapse
Affiliation(s)
- Fenglin Zhang
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Jingliang Ye
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Junle Zhu
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Wenbo Qian
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Haoheng Wang
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Chun Luo
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
21
|
Ghosh S, Bhaskar R, Mishra R, Arockia Babu M, Abomughaid MM, Jha NK, Sinha JK. Neurological insights into brain-targeted cancer therapy and bioinspired microrobots. Drug Discov Today 2024; 29:104105. [PMID: 39029869 DOI: 10.1016/j.drudis.2024.104105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024]
Abstract
Cancer, a multifaceted and pernicious disease, continuously challenges medicine, requiring innovative treatments. Brain cancers pose unique and daunting challenges due to the intricacies of the central nervous system and the blood-brain barrier. In this era of precision medicine, the convergence of neurology, oncology, and cutting-edge technology has given birth to a promising avenue - targeted cancer therapy. Furthermore, bioinspired microrobots have emerged as an ingenious approach to drug delivery, enabling precision and control in cancer treatment. This Keynote review explores the intricate web of neurological insights into brain-targeted cancer therapy and the paradigm-shifting world of bioinspired microrobots. It serves as a critical and comprehensive overview of these evolving fields, aiming to underscore their integration and potential for revolutionary cancer treatments.
Collapse
Affiliation(s)
- Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Richa Mishra
- Department of Computer Science and Engineering, Parul University, Vadodara, Gujrat 391760, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Niraj Kumar Jha
- Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | | |
Collapse
|
22
|
Piao C, Lee J, Kim GE, Choe YH, Lee H, Hyun YM. Targeted Delivery of Nanoparticle-Conveyed Neutrophils to the Glioblastoma Site for Efficient Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:41819-41827. [PMID: 39057192 PMCID: PMC11332397 DOI: 10.1021/acsami.4c05691] [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: 04/08/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Glioblastoma is a common brain tumor that poses considerable challenges in drug delivery. In this study, we investigated the potential of cell-based nanoparticles for targeted drug delivery to the glioblastoma sites. The anticancer drug of temozolomide (TMZ)-loaded T7-cholesterol nanoparticle micelles efficiently delivered nanoparticles to neutrophils and, subsequently, to the tumors. T7 is a cell-penetrating peptide that enhances the delivery of T7/TMZ to the target cells. T7 also serves as a transferrin target peptide, enabling targeted delivery to tumors. T7-conjugated cholesterol can self-assemble into micelles in aqueous solution and attach to the membrane of neutrophils. We confirmed that T7/TMZ nanoparticle micelles were efficiently located inside the neutrophils. Thereafter, T7/TMZ-conveyed neutrophils were administered to a glioblastoma mouse model, enabling neutrophils to penetrate the blood-brain barrier and deliver drugs directly to the tumor site. We evaluated the drug delivery efficiency and therapeutic effects of intravenous injection of T7/TMZ-conveyed neutrophils to a glioblastoma mouse model. These results demonstrate the promising role of neutrophil-based nanoparticle delivery systems in the targeted therapy of glioblastoma.
Collapse
Affiliation(s)
- Chunxian Piao
- Department
of Anatomy, Yonsei University College of
Medicine, Seoul 03722, Republic of Korea
| | - Jaeho Lee
- Department
of Anatomy, Yonsei University College of
Medicine, Seoul 03722, Republic of Korea
- Brain
Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic
of Korea
| | - Gi Eun Kim
- Department
of Anatomy, Yonsei University College of
Medicine, Seoul 03722, Republic of Korea
- Brain
Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic
of Korea
| | - Young Ho Choe
- Department
of Anatomy, Yonsei University College of
Medicine, Seoul 03722, Republic of Korea
- Brain
Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic
of Korea
| | - Haerang Lee
- Department
of Anatomy, Yonsei University College of
Medicine, Seoul 03722, Republic of Korea
- Brain
Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic
of Korea
| | - Young-Min Hyun
- Department
of Anatomy, Yonsei University College of
Medicine, Seoul 03722, Republic of Korea
- Brain
Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic
of Korea
| |
Collapse
|
23
|
Wu M, Wang T, Ji N, Lu T, Yuan R, Wu L, Zhang J, Li M, Cao P, Zhao J, Li G, Li J, Li Y, Tang Y, Gao Z, Wang X, Cheng W, Ge M, Cui G, Li R, Wu A, You Y, Zhang W, Wang Q, Chen J. Multi-omics and pharmacological characterization of patient-derived glioma cell lines. Nat Commun 2024; 15:6740. [PMID: 39112531 PMCID: PMC11306361 DOI: 10.1038/s41467-024-51214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Glioblastoma (GBM) is the most common brain tumor and remains incurable. Primary GBM cultures are widely used tools for drug screening, but there is a lack of genomic and pharmacological characterization for these primary GBM cultures. Here, we collect 50 patient-derived glioma cell (PDGC) lines and characterize them by whole genome sequencing, RNA sequencing, and drug response screening. We identify three molecular subtypes among PDGCs: mesenchymal (MES), proneural (PN), and oxidative phosphorylation (OXPHOS). Drug response profiling reveals that PN subtype PDGCs are sensitive to tyrosine kinase inhibitors, whereas OXPHOS subtype PDGCs are sensitive to histone deacetylase inhibitors, oxidative phosphorylation inhibitors, and HMG-CoA reductase inhibitors. PN and OXPHOS subtype PDGCs stably form tumors in vivo upon intracranial transplantation into immunodeficient mice, whereas most MES subtype PDGCs fail to form tumors in vivo. In addition, PDGCs cultured by serum-free medium, especially long-passage PDGCs, carry MYC/MYCN amplification, which is rare in GBM patients. Our study provides a valuable resource for understanding primary glioma cell cultures and clinical translation and highlights the problems of serum-free PDGC culture systems that cannot be ignored.
Collapse
Affiliation(s)
- Min Wu
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, China
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tingting Wang
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Changping Laboratory, Beijing, China
- Chinese Institute for Brain Research, Beijing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ting Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ran Yuan
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Junxia Zhang
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengyuan Li
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Changping Laboratory, Beijing, China
| | - Penghui Cao
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiarui Zhao
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Guanzhang Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jianyu Li
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Li
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujie Tang
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengliang Gao
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Department of Anesthesiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Xiuxing Wang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming Ge
- Department of Neurosurgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Rui Li
- Department of Neurosurgery, China-Japan Friendship Hospital, No. 2 Yinghua East Road, Chaoyang District, Beijing, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongping You
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China.
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Jian Chen
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China.
- Changping Laboratory, Beijing, China.
- Chinese Institute for Brain Research, Beijing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Budhiraja S, McManus G, Baisiwala S, Perrault EN, Cho S, Saathoff M, Chen L, Park CH, Kazi HA, Dmello C, Lin P, James CD, Sonabend AM, Heiland DH, Ahmed AU. ARF4-mediated retrograde trafficking as a driver of chemoresistance in glioblastoma. Neuro Oncol 2024; 26:1421-1437. [PMID: 38506351 PMCID: PMC11300013 DOI: 10.1093/neuonc/noae059] [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] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Cellular functions hinge on the meticulous orchestration of protein transport, both spatially and temporally. Central to this process is retrograde trafficking, responsible for targeting proteins to the nucleus. Despite its link to many diseases, the implications of retrograde trafficking in glioblastoma (GBM) are still unclear. METHODS To identify genetic drivers of TMZ resistance, we conducted comprehensive CRISPR-knockout screening, revealing ADP-ribosylation factor 4 (ARF4), a regulator of retrograde trafficking, as a major contributor. RESULTS Suppressing ARF4 significantly enhanced TMZ sensitivity in GBM patient-derived xenograft (PDX) models, leading to improved survival rates (P < .01) in both primary and recurrent lines. We also observed that TMZ exposure stimulates ARF4-mediated retrograde trafficking. Proteomics analysis of GBM cells with varying levels of ARF4 unveiled the influence of this pathway on EGFR signaling, with increased nuclear trafficking of EGFR observed in cells with ARF4 overexpression and TMZ treatment. Additionally, spatially resolved RNA-sequencing of GBM patient tissues revealed substantial correlations between ARF4 and crucial nuclear EGFR (nEGFR) downstream targets, such as MYC, STAT1, and DNA-PK. Decreased activity of DNA-PK, a DNA repair protein downstream of nEGFR signaling that contributes to TMZ resistance, was observed in cells with suppressed ARF4 levels. Notably, treatment with DNA-PK inhibitor, KU-57788, in mice with a recurrent PDX line resulted in prolonged survival (P < .01), highlighting the promising therapeutic implications of targeting proteins reliant on ARF4-mediated retrograde trafficking. CONCLUSIONS Our findings demonstrate that ARF4-mediated retrograde trafficking contributes to the development of TMZ resistance, cementing this pathway as a viable strategy to overcome chemoresistance in GBM.
Collapse
Affiliation(s)
- Shreya Budhiraja
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Graysen McManus
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Ella N Perrault
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sia Cho
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Miranda Saathoff
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Cheol H Park
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hasaan A Kazi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peiyu Lin
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dieter H Heiland
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Microenvironment and Immunology Research Laboratory, Medical Center - University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Freiburg, Germany
| | - Atique U Ahmed
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
25
|
Pinyon JL, von Jonquieres G, Crawford EN, Abed AA, Power JM, Klugmann M, Browne CJ, Housley DM, Wise AK, Fallon JB, Shepherd RK, Lin JY, McMahon C, McAlpine D, Birman CS, Lai W, Enke YL, Carter PM, Patrick JF, Gay RD, Marie C, Scherman D, Lovell NH, Housley GD. Gene Electrotransfer via Conductivity-Clamped Electric Field Focusing Pivots Sensori-Motor DNA Therapeutics: "A Spoonful of Sugar Helps the Medicine Go Down". ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401392. [PMID: 38874431 PMCID: PMC11321635 DOI: 10.1002/advs.202401392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/20/2024] [Indexed: 06/15/2024]
Abstract
Viral vectors and lipofection-based gene therapies have dispersion-dependent transduction/transfection profiles that thwart precise targeting. The study describes the development of focused close-field gene electrotransfer (GET) technology, refining spatial control of gene expression. Integration of fluidics for precise delivery of "naked" plasmid deoxyribonucleic acid (DNA) in sucrose carrier within the focused electric field enables negative biasing of near-field conductivity ("conductivity-clamping"-CC), increasing the efficiency of plasma membrane molecular translocation. This enables titratable gene delivery with unprecedently low charge transfer. The clinic-ready bionics-derived CC-GET device achieved neurotrophin-encoding miniplasmid DNA delivery to the cochlea to promote auditory nerve regeneration; validated in deafened guinea pig and cat models, leading to improved central auditory tuning with bionics-based hearing. The performance of CC-GET is evaluated in the brain, an organ problematic for pulsed electric field-based plasmid DNA delivery, due to high required currents causing Joule-heating and damaging electroporation. Here CC-GET enables safe precision targeting of gene expression. In the guinea pig, reporter expression is enabled in physiologically critical brainstem regions, and in the striatum (globus pallidus region) delivery of a red-shifted channelrhodopsin and a genetically-encoded Ca2+ sensor, achieved photoactivated neuromodulation relevant to the treatment of Parkinson's Disease and other focal brain disorders.
Collapse
Affiliation(s)
- Jeremy L. Pinyon
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
- Charles Perkins CentreSchool of Medical SciencesFaculty of Medicine and HealthUniversity of SydneySydneyNSW2006Australia
| | - Georg von Jonquieres
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Edward N. Crawford
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Amr Al Abed
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - John M. Power
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Matthias Klugmann
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Cherylea J. Browne
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
- Medical SciencesSchool of ScienceWestern Sydney UniversitySydneyNSW2560Australia
| | - David M. Housley
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Andrew K. Wise
- Bionics Institute384–388 Albert StreetEast MelbourneVIC3002Australia
- Medical BionicsDepartment of OtolaryngologyUniversity of MelbourneMelbourneVIC3002Australia
| | - James B. Fallon
- Bionics Institute384–388 Albert StreetEast MelbourneVIC3002Australia
- Medical BionicsDepartment of OtolaryngologyUniversity of MelbourneMelbourneVIC3002Australia
| | - Robert K. Shepherd
- Bionics Institute384–388 Albert StreetEast MelbourneVIC3002Australia
- Medical BionicsDepartment of OtolaryngologyUniversity of MelbourneMelbourneVIC3002Australia
| | - John Y. Lin
- Tasmanian School of MedicineUniversity of TasmaniaHobartTAS7001Australia
| | - Catherine McMahon
- Faculty of Medicine and Health SciencesThe Hearing HubMacquarie UniversitySydney2109Australia
| | - David McAlpine
- Faculty of Medicine and Health SciencesThe Hearing HubMacquarie UniversitySydney2109Australia
| | - Catherine S. Birman
- Faculty of Medicine and Health SciencesThe Hearing HubMacquarie UniversitySydney2109Australia
- Faculty of Medicine and HealthUniversity of SydneySydneyNSW2006Australia
- Department of OtolaryngologyRoyal Prince Alfred HospitalCamperdownNSW2050Australia
- NextSenseRoyal Institute of Deaf and Blind ChildrenGladesvilleNSW2111Australia
| | - Waikong Lai
- NextSenseRoyal Institute of Deaf and Blind ChildrenGladesvilleNSW2111Australia
| | - Ya Lang Enke
- Cochlear LimitedMacquarie UniversityUniversity AvenueMacquarie ParkNSW2109Australia
| | - Paul M. Carter
- Cochlear LimitedMacquarie UniversityUniversity AvenueMacquarie ParkNSW2109Australia
| | - James F. Patrick
- Cochlear LimitedMacquarie UniversityUniversity AvenueMacquarie ParkNSW2109Australia
| | - Robert D. Gay
- Cochlear LimitedMacquarie UniversityUniversity AvenueMacquarie ParkNSW2109Australia
| | - Corinne Marie
- CNRS, Inserm, UTCBSUniversité Paris CitéParisF‐75006France
- Chimie ParisTechUniversité PSLParis75005France
| | - Daniel Scherman
- CNRS, Inserm, UTCBSUniversité Paris CitéParisF‐75006France
- Fondation Maladies Rares96 rue DidotParis75014France
| | - Nigel H. Lovell
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| | - Gary D. Housley
- Translational Neuroscience FacilityDepartment of PhysiologySchool of Biomedical SciencesGraduate School of Biomedical EngineeringTyree Institute for Health Engineering (IHealthE)UNSWSydneyNSW2052Australia
| |
Collapse
|
26
|
Zhang X, Shao X, Bao Q, He L, Qi X. Integrated network pharmacology and experimental verification to reveal the role of Shezhi Huangling Decoction against glioma by inactivating PI3K/Akt-HIF1A axis. Heliyon 2024; 10:e34215. [PMID: 39092253 PMCID: PMC11292238 DOI: 10.1016/j.heliyon.2024.e34215] [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: 06/04/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Shezhi Huangling Decoction (SHD) has been proven clinically effective in regulating metabolic and immune homeostasis in the treatment of glioma. The investigation aimed to deconstruct the active constituents and mechanisms of SHD. Effects of SHD on malignant characteristics of HS683 and KNS89 cells have been investigated by CCK-8, clone formation, flow cytometry, and Transwell assays. A mouse xenograft model was established to assess the effect of SHD or SHD + temozolomide (TMZ) in vivo. A total of 461 constituents were found from SHD in UPLC/Q-TOF-MS/MS analysis. Functional enrichment analysis showed that pathway in cancer, proteoglycans in cancer, regulation of epithelial cell proliferation, inflammation/immune, gliogenesis, brain development, cell adhesion, and autophagy could participate in the treatment of SHD. Additionally, 9 hub genes (AKT1, TP53, CTNNB1, STAT3, EGFR, VEGFA, PIK3CA, ERBB2, and HIF1A) were identified as hub genes. Moreover, we found that SHD may greatly reduce the migration and accelerate apoptosis of HS683 and KNS89 cells. Additionally, SHD coordinates TMZ to restrict tumor growth were found in the mice. Our results suggest that the malignant behaviors of glioma cells are suppressed by SHD and the mechanism may be closing on the inhibition of the PI3K/Akt-HIF1A axis. SHD may serve as a synergistic therapeutic choice for TMZ to suppress glioblastoma growth.
Collapse
Affiliation(s)
- Xiaobing Zhang
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Xian Shao
- Department of Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Qingquan Bao
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Lingyan He
- Department of Traditional Chinese Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
27
|
Sarkar S, Greer J, Marlowe NJ, Medvid A, Ivan ME, Kolishetti N, Dhar S. Stemness, invasion, and immunosuppression modulation in recurrent glioblastoma using nanotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1976. [PMID: 39091260 DOI: 10.1002/wnan.1976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 08/04/2024]
Abstract
The recurrent nature of glioblastoma negatively impacts conventional treatment strategies leading to a growing need for nanomedicine. Nanotherapeutics, an approach designed to deliver drugs to specific sites, is experiencing rapid growth and gaining immense popularity. Having potential in reaching the hard-to-reach disease sites, this field has the potential to show high efficacy in combatting glioblastoma progression. The presence of glioblastoma stem cells (GSCs) is a major factor behind the poor prognosis of glioblastoma multiforme (GBM). Stemness potential, heterogeneity, and self-renewal capacity, are some of the properties that make GSCs invade across the distant regions of the brain. Despite advances in medical technology and MRI-guided maximal surgical resection, not all GSCs residing in the brain can be removed, leading to recurrent disease. The aggressiveness of GBM is often correlated with immune suppression, where the T-cells are unable to infiltrate the cancer initiating GSCs. Standard of care therapies, including surgery and chemotherapy in combination with radiation therapy, have failed to tackle all the challenges of the GSCs, making it increasingly important for researchers to develop strategies to tackle their growth and proliferation and reduce the recurrence of GBM. Here, we will focus on the advancements in the field of nanomedicine that has the potential to show positive impact in managing glioblastoma tumor microenvironment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Shrita Sarkar
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Jessica Greer
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Nathaniel J Marlowe
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Angeline Medvid
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Michael E Ivan
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Institute of Neuroimmune Pharmacology, Miami, Florida International University, Florida, USA
| | - Shanta Dhar
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Chemistry, University of Miami, Coral Gables, Florida, USA
| |
Collapse
|
28
|
Ghadami E, Gorji A, Pour-Rashidi A, Noorbakhsh F, Kabuli M, Razipour M, Choobineh H, Maghsudlu M, Damavandi E, Ghadami M. CircZNF609 and circNFIX as possible regulators of glioblastoma pathogenesis via miR-145-5p/EGFR axis. Sci Rep 2024; 14:13551. [PMID: 38866807 PMCID: PMC11169398 DOI: 10.1038/s41598-024-63827-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
Glioblastoma is a rare and deadly malignancy with a low survival rate. Emerging evidence has shown that aberrantly expressed circular RNAs (circRNAs) play a critical role in the initiation and progression of GBM tumorigenesis. The oncogenic function of circZNF609 and circNFIX is involved in several types of cancer, but the role and underlying mechanism of these circRNAs in glioblastoma remain unclear. In this study, we hypothesized that circZNF609 and circNFIX may regulate EGFR through sponging miR-145-5p. Herein, we assessed the expression levels of circZNF609, circNFIX, miR-145-5p, and EGFR using quantitative polymerase chain reaction in glioblastoma patients and normal brain samples. The results showed that circZNF609, circNFIX, and EGFR expression levels were upregulated and miR145-5p was downregulated (p = 0.001, 0.06, 0.002, and 0.0065, respectively), while there was no significant association between clinicopathological features of the patients and the level of these genes expression. We also found a significant inverse correlation between miR145-5p and the expression of cZNF609, cNFIX and EGFR (p = 0.0003, 0.0006, and 0.009, respectively). These findings may open a new window for researchers to better understand the potential pathways involved in GBM pathogenesis. In conclusion, it may provide a new potential pathway for the development of effective drugs for the treatment of GBM patients.
Collapse
Affiliation(s)
- Elham Ghadami
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Al-Anbia Hospital, Tehran, Iran
- Department of Neurosurgery, Westfälische Wilhelms-Universität, Munster, Germany
| | - Ahmad Pour-Rashidi
- Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Kabuli
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Razipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Choobineh
- School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohaddese Maghsudlu
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elia Damavandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
- Specialized Medical Genetic Center (SMGC) of ACECR, Tehran, Iran
| | - Mohsen Ghadami
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cardiac Primary Research Center, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.
- Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Zhou Z, Leng H. Deciphering the causal relationship between plasma and cerebrospinal fluid metabolites and glioblastoma multiforme: a Mendelian Randomization study. Aging (Albany NY) 2024; 16:8306-8319. [PMID: 38742944 PMCID: PMC11131984 DOI: 10.18632/aging.205818] [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: 12/26/2023] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Glioblastoma Multiforme (GBM) is one of the most aggressive and fatal brain cancers. The study of metabolites could be crucial for understanding GBM's biology and reveal new treatment strategies. METHODS The GWAS data for GBM were sourced from the FinnGen database. A total of 1400 plasma metabolites were collected from the GWAS Catalog dataset. The cerebrospinal fluid (CSF) metabolites data were collected from subsets of participants in the WADRC and WRAP studies. We utilized the inverse variance weighting (IVW) method as the primary tool to explore the causal relationship between metabolites in plasma and CSF and glioblastoma, ensuring the exclusion of instances with horizontal pleiotropy. Additionally, four supplementary analytical methods were applied to reinforce our findings. Aberrant results were identified and omitted based on the outcomes of the leave-one-out sensitivity analysis. Conclusively, a reverse Mendelian Randomization analysis was also conducted to further substantiate our results. RESULTS The study identified 69 plasma metabolites associated with GBM. Of these, 40 metabolites demonstrated a significant positive causal relationship with GBM, while 29 exhibited a significant negative causal association. Notably, Trimethylamine N-oxide (TMAO) levels in plasma, not CSF, were found to be a significant exposure factor for GBM (OR = 3.1627, 95% CI = (1.6347, 6.1189), P = 0.0006). The study did not find a reverse causal relationship between GBM and plasma TMAO levels. CONCLUSIONS This research has identified 69 plasma metabolites potentially associated with the incidence of GBM, among which TMAO stands out as a promising candidate for an early detectable biomarker for GBM.
Collapse
Affiliation(s)
- Zhiwei Zhou
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan 415003, People’s Republic of China
| | - Haibin Leng
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan 415003, People’s Republic of China
| |
Collapse
|
30
|
Lim SH, Yee GT, Khang D. Nanoparticle-Based Combinational Strategies for Overcoming the Blood-Brain Barrier and Blood-Tumor Barrier. Int J Nanomedicine 2024; 19:2529-2552. [PMID: 38505170 PMCID: PMC10949308 DOI: 10.2147/ijn.s450853] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024] Open
Abstract
The blood-brain barrier (BBB) and blood-tumor barrier (BTB) pose substantial challenges to efficacious drug delivery for glioblastoma multiforme (GBM), a primary brain tumor with poor prognosis. Nanoparticle-based combinational strategies have emerged as promising modalities to overcome these barriers and enhance drug penetration into the brain parenchyma. This review discusses various nanoparticle-based combinatorial approaches that combine nanoparticles with cell-based drug delivery, viral drug delivery, focused ultrasound, magnetic field, and intranasal drug delivery to enhance drug permeability across the BBB and BTB. Cell-based drug delivery involves using engineered cells as carriers for nanoparticles, taking advantage of their intrinsic migratory and homing capabilities to facilitate the transport of therapeutic payloads across BBB and BTB. Viral drug delivery uses engineered viral vectors to deliver therapeutic genes or payloads to specific cells within the GBM microenvironment. Focused ultrasound, coupled with microbubbles or nanoparticles, can temporarily disrupt the BBB to increase drug permeability. Magnetic field-guided drug delivery exploits magnetic nanoparticles to facilitate targeted drug delivery under an external magnetic field. Intranasal drug delivery offers a minimally invasive avenue to bypass the BBB and deliver therapeutic agents directly to the brain via olfactory and trigeminal pathways. By combining these strategies, synergistic effects can enhance drug delivery efficiency, improve therapeutic efficacy, and reduce off-target effects. Future research should focus on optimizing nanoparticle design, exploring new combination strategies, and advancing preclinical and clinical investigations to promote the translation of nanoparticle-based combination therapies for GBM.
Collapse
Affiliation(s)
- Su Hyun Lim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Gi Taek Yee
- Department of Neurosurgery, Gil Medical Center, Gachon University, School of Medicine, Incheon, 21565, South Korea
| | - Dongwoo Khang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
| |
Collapse
|
31
|
Liao C, Chen W, Xu G, Wang J, Dong W. High expression of ARPC1B correlates with immune infiltration and poor outcomes in glioblastoma. Biochem Biophys Rep 2024; 37:101619. [PMID: 38188361 PMCID: PMC10770598 DOI: 10.1016/j.bbrep.2023.101619] [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: 10/10/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024] Open
Abstract
Objective To investigate the role of ARPC1B in GBM and its prognostic value. Methods mRNA and protein expression of ARPC1B in GBM was analyzed using the TCGA; TIMER2 and the HPA databases, and protein expression differences were detected using immunohistochemistry. K-M analysis and Cox regression analysis were performed on high and low ARPC1B expression groups in the TCGA database. The relationship between immune cells and ARPC1B expression was explored using the TIMER2 database. GO and KEGG analyses were conducted to investigate the functions of ARPC1B-related genes in GBM. Results ARPC1B was highly expressed in both GBM tissues and cell lines, and it was demonstrated as a prognostic biomarker for GBM. ARPC1B expression levels showed associations with immune cell populations within the GBM microenvironment. Conclusion ARPC1B can regulating immune infiltration in the GBM microenvironment, indicating its potential as a novel therapeutic target for GBM.
Collapse
Affiliation(s)
- Chuangxin Liao
- Corresponding author. Department of Neurosurgery, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-sen University, 183 Huangpu Road East, Guangzhou, 510700, China.
| | | | | | - Jingshan Wang
- Department of Neurosurgery, The Eastern Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijie Dong
- Department of Neurosurgery, The Eastern Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
32
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [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: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
33
|
Habeeb M, Vengateswaran HT, You HW, Saddhono K, Aher KB, Bhavar GB. Nanomedicine facilitated cell signaling blockade: difficulties and strategies to overcome glioblastoma. J Mater Chem B 2024; 12:1677-1705. [PMID: 38288615 DOI: 10.1039/d3tb02485g] [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: 02/15/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive and lethal type of brain tumor with complex and diverse molecular signaling pathways involved that are in its development and progression. Despite numerous attempts to develop effective treatments, the survival rate remains low. Therefore, understanding the molecular mechanisms of these pathways can aid in the development of targeted therapies for the treatment of glioblastoma. Nanomedicines have shown potential in targeting and blocking signaling pathways involved in glioblastoma. Nanomedicines can be engineered to specifically target tumor sites, bypass the blood-brain barrier (BBB), and release drugs over an extended period. However, current nanomedicine strategies also face limitations, including poor stability, toxicity, and low therapeutic efficacy. Therefore, novel and advanced nanomedicine-based strategies must be developed for enhanced drug delivery. In this review, we highlight risk factors and chemotherapeutics for the treatment of glioblastoma. Further, we discuss different nanoformulations fabricated using synthetic and natural materials for treatment and diagnosis to selectively target signaling pathways involved in GBM. Furthermore, we discuss current clinical strategies and the role of artificial intelligence in the field of nanomedicine for targeting GBM.
Collapse
Affiliation(s)
- Mohammad Habeeb
- Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai-600048, India.
| | - Hariharan Thirumalai Vengateswaran
- Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai-600048, India.
| | - Huay Woon You
- Pusat PERMATA@Pintar Negara, Universiti Kebangsaan 43600, Bangi, Selangor, Malaysia
| | - Kundharu Saddhono
- Faculty of Teacher Training and Education, Universitas Sebelas Maret, 57126, Indonesia
| | - Kiran Balasaheb Aher
- Department of Pharmaceutical Quality Assurance, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - Girija Balasaheb Bhavar
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| |
Collapse
|
34
|
Zhao C, Zhu X, Tan J, Mei C, Cai X, Kong F. Lipid-based nanoparticles to address the limitations of GBM therapy by overcoming the blood-brain barrier, targeting glioblastoma stem cells, and counteracting the immunosuppressive tumor microenvironment. Biomed Pharmacother 2024; 171:116113. [PMID: 38181717 DOI: 10.1016/j.biopha.2023.116113] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor, characterized by high heterogeneity, strong invasiveness, poor prognosis, and a low survival rate. A broad range of nanoparticles have been recently developed as drug delivery systems for GBM therapy owing to their inherent size effect and ability to cross the blood-brain barrier (BBB). Lipid-based nanoparticles (LBNPs), such as liposomes, solid lipid NPs (SLNs), and nano-structured lipid carriers (NLCs), have emerged as the most promising drug delivery system for the treatment of GBM because of their unique size, surface modification possibilities, and proven bio-safety. In this review, the main challenges of the current clinical treatment of GBM and the strategies on how novel LBNPs overcome them were explored. The application and progress of LBNP-based drug delivery systems in GBM chemotherapy, immunotherapy, and gene therapy in recent years were systematically reviewed, and the prospect of LBNPs for GBM treatment was discussed.
Collapse
Affiliation(s)
- Changhong Zhao
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China; Lantian Pharmaceuticals Co., Ltd, Hubei, China.
| | - Xinshu Zhu
- School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an 223005, China
| | - Jianmei Tan
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Chao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Xiang Cai
- Lantian Pharmaceuticals Co., Ltd, Hubei, China; School of Business, Hubei University of Science and Technology, China
| | - Fei Kong
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China; School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
35
|
Nóbrega AHL, Pimentel RS, Prado AP, Garcia J, Frozza RL, Bernardi A. Neuroinflammation in Glioblastoma: The Role of the Microenvironment in Tumour Progression. Curr Cancer Drug Targets 2024; 24:579-594. [PMID: 38310461 DOI: 10.2174/0115680096265849231031101449] [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: 06/30/2023] [Revised: 08/25/2023] [Accepted: 09/08/2023] [Indexed: 02/05/2024]
Abstract
Glioblastoma (GBM) stands as the most aggressive and lethal among the main types of primary brain tumors. It exhibits malignant growth, infiltrating the brain tissue, and displaying resistance toward treatment. GBM is a complex disease characterized by high degrees of heterogeneity. During tumour growth, microglia and astrocytes, among other cells, infiltrate the tumour microenvironment and contribute extensively to gliomagenesis. Tumour-associated macrophages (TAMs), either of peripheral origin or representing brain-intrinsic microglia, are the most numerous nonneoplastic populations in the tumour microenvironment in GBM. The complex heterogeneous nature of GBM cells is facilitated by the local inflammatory tumour microenvironment, which mostly induces tumour aggressiveness and drug resistance. The immunosuppressive tumour microenvironment of GBM provides multiple pathways for tumour immune evasion, contributing to tumour progression. Additionally, TAMs and astrocytes can contribute to tumour progression through the release of cytokines and activation of signalling pathways. In this review, we summarize the role of the microenvironment in GBM progression, focusing on neuroinflammation. These recent advancements in research of the microenvironment hold the potential to offer a promising approach to the treatment of GBM in the coming times.
Collapse
Affiliation(s)
| | - Rafael Sampaio Pimentel
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Ana Paula Prado
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Jenifer Garcia
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Rudimar Luiz Frozza
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Andressa Bernardi
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| |
Collapse
|
36
|
Khan RB, Tiwari S, Jarkharya A, Tiwari A, Chowdhary R, Shrivastava A. Glioblastoma Multiforme miRNA based Comprehensive Study to Validate Phytochemicals for Effective Treatment against Deadly Tumour through In Silico Evaluation. Microrna 2024; 13:240-250. [PMID: 38982916 DOI: 10.2174/0122115366302365240618122812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Glioblastoma Multiforme (GBM) is a prevalent and deadly type of primary astrocytoma, constituting over 60% of adult brain tumors, and has a poor prognosis, with a high relapse rate within 7 months of diagnosis. Despite surgical, radiotherapy, and chemotherapy treatments, GBM remains challenging due to resistance. MicroRNA (miRNAs) control gene expression at transcriptional and post-transcriptional levels by targeting their messenger RNA (mRNA), and also contribute to the development of various neoplasms, including GBM. METHODS The present study focuses on exploring the miRNAs-based pathogenesis of GBM and evaluating most potential plant-based therapeutic agents with in silico analysis. Gene chips were retrieved from the Gene Expression Omnibus (GEO) database, followed by the Robust- Rank- Aggereg algorithm to determine the Differentially Expressed miRNAs (DEMs). The predicted targets were intersected with the GBM-associated genes, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of the overlapping genes was performed. At the same time, five phytochemicals were selected for the Connectivity map (CMap), and the most efficient ones were those that had undergone molecular docking analysis to obtain the potential therapeutic agents. RESULTS The hsa-miR-10b, hsa-miR-21, and hsa-miR-15b were obtained, and eight genes were found to be associated with glioma pathways; VSIG4, PROCR, PLAT, and ITGB2 were upregulated while, CAMK2B, PDE1A, GABRA1, and KCNJ6 were downregulated. The drugs Resveratrol and Quercetin were identified as the most prominent drugs. CONCLUSION These miRNAs-based drugs can be used as a curative agent for the treatment of GBM. However, in vivo, experimental data, and clinical trials are necessary to provide an alternative to conventional GBM cancer chemotherapy.
Collapse
Affiliation(s)
- Roji Begam Khan
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India
- School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya Bhopal, 462036, India
| | - Shikha Tiwari
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India
| | - Aryan Jarkharya
- School of Biological Sciences and Biotechnology, Goa University, Taliegaon Plataeu, Bambolim, Goa, 403206, India
| | - Archana Tiwari
- School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya Bhopal, 462036, India
| | - Rashmi Chowdhary
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India
| | - Adesh Shrivastava
- Department of Neurosurgery, All India Institute of Medical Sciences Bhopal, Bhopal, India
| |
Collapse
|
37
|
Kusaczuk M, Ambel ET, Naumowicz M, Velasco G. Cellular stress responses as modulators of drug cytotoxicity in pharmacotherapy of glioblastoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189054. [PMID: 38103622 DOI: 10.1016/j.bbcan.2023.189054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Despite the extensive efforts to find effective therapeutic strategies, glioblastoma (GBM) remains a therapeutic challenge with dismal prognosis of survival. Over the last decade the role of stress responses in GBM therapy has gained a great deal of attention, since depending on the duration and intensity of these cellular programs they can be cytoprotective or promote cancer cell death. As such, initiation of the UPR, autophagy or oxidative stress may either impede or facilitate drug-mediated cell killing. In this review, we summarize the mechanisms that regulate ER stress, autophagy, and oxidative stress during GBM development and progression to later discuss the involvement of these stress pathways in the response to different treatments. We also discuss how a precise understanding of the molecular mechanisms regulating stress responses evoked by different pharmacological agents could decisively contribute to the design of novel and more effective combinational treatments against brain malignancies.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland.
| | - Elena Tovar Ambel
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain
| | - Monika Naumowicz
- Department of Physical Chemistry, Faculty of Chemistry, University of Bialystok, K. Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain.
| |
Collapse
|
38
|
Singh S, Joshi V, Upadhyay A. Amyloids and brain cancer: molecular linkages and crossovers. Biosci Rep 2023; 43:BSR20230489. [PMID: 37335084 PMCID: PMC10548166 DOI: 10.1042/bsr20230489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
Amyloids are high-order proteinaceous formations deposited in both intra- and extracellular spaces. These aggregates have tendencies to deregulate cellular physiology in multiple ways; for example, altered metabolism, mitochondrial dysfunctions, immune modulation, etc. When amyloids are formed in brain tissues, the endpoint often is death of neurons. However, interesting but least understood is a close connection of amyloids with another set of conditions in which brain cells proliferate at an extraordinary rate and form tumor inside brain. Glioblastoma is one such condition. Increasing number of evidence indicate a possible link between amyloid formation and depositions in brain tumors. Several proteins associated with cell cycle regulation and apoptotic pathways themselves have shown to possess high tendencies to form amyloids. Tumor suppressor protein p53 is one prominent example that mutate, oligomerize and form amyloids leading to loss- or gain-of-functions and cause increased cell proliferation and malignancies. In this review article, we present available examples, genetic links and common pathways that indicate that possibly the two distantly placed pathways: amyloid formation and developing cancers in the brain have similarities and are mechanistically intertwined together.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| | - Vibhuti Joshi
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh 201310, India
| | - Arun Upadhyay
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| |
Collapse
|
39
|
Rabah N, Ait Mohand FE, Kravchenko-Balasha N. Understanding Glioblastoma Signaling, Heterogeneity, Invasiveness, and Drug Delivery Barriers. Int J Mol Sci 2023; 24:14256. [PMID: 37762559 PMCID: PMC10532387 DOI: 10.3390/ijms241814256] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The most prevalent and aggressive type of brain cancer, namely, glioblastoma (GBM), is characterized by intra- and inter-tumor heterogeneity and strong spreading capacity, which makes treatment ineffective. A true therapeutic answer is still in its infancy despite various studies that have made significant progress toward understanding the mechanisms behind GBM recurrence and its resistance. The primary causes of GBM recurrence are attributed to the heterogeneity and diffusive nature; therefore, monitoring the tumor's heterogeneity and spreading may offer a set of therapeutic targets that could improve the clinical management of GBM and prevent tumor relapse. Additionally, the blood-brain barrier (BBB)-related poor drug delivery that prevents effective drug concentrations within the tumor is discussed. With a primary emphasis on signaling heterogeneity, tumor infiltration, and computational modeling of GBM, this review covers typical therapeutic difficulties and factors contributing to drug resistance development and discusses potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem 91120, Israel; (N.R.); (F.-E.A.M.)
| |
Collapse
|
40
|
Nowak I, Madej M, Secemska J, Sarna R, Strzalka-Mrozik B. Virus-Based Biological Systems as Next-Generation Carriers for the Therapy of Central Nervous System Diseases. Pharmaceutics 2023; 15:1931. [PMID: 37514117 PMCID: PMC10384784 DOI: 10.3390/pharmaceutics15071931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Central nervous system (CNS) diseases are currently a major challenge in medicine. One reason is the presence of the blood-brain barrier, which is a significant limitation for currently used medicinal substances that are characterized by a high molecular weight and a short half-life. Despite the application of nanotechnology, there is still the problem of targeting and the occurrence of systemic toxicity. Viral vectors and virus-like particles (VLPs) may provide a promising solution to these challenges. Their small size, biocompatibility, ability to carry medicinal substances, and specific targeting of neural cells make them useful in research when formulating a new generation of biological carriers. Additionally, the possibility of genetic modification has the potential for gene therapy. Among the most promising viral vectors are adeno-associated viruses, adenoviruses, and retroviruses. This is due to their natural tropism to neural cells, as well as the possibility of genetic and surface modification. Moreover, VLPs that are devoid of infectious genetic material in favor of increasing capacity are also leading the way for research on new drug delivery systems. The aim of this study is to review the most recent reports on the use of viral vectors and VLPs in the treatment of selected CNS diseases.
Collapse
Affiliation(s)
- Ilona Nowak
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Marcel Madej
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Julia Secemska
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Robert Sarna
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Barbara Strzalka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
41
|
Liu X, Sun C, Wang Q, Li P, Zhao T, Li Q. Sp1 Upregulation Bolsters the Radioresistance of Glioblastoma Cells by Promoting Double Strand Breaks Repair. Int J Mol Sci 2023; 24:10658. [PMID: 37445835 PMCID: PMC10342049 DOI: 10.3390/ijms241310658] [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: 05/18/2023] [Revised: 06/08/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Radioresistance remains a critical obstacle in the clinical management of glioblastoma (GBM) by radiotherapy. Therefore, it is necessary to explore the molecular mechanisms underlying radioresistance to improve patient response to radiotherapy and increase the treatment efficacy. The present study aimed to elucidate the role of specificity protein 1 (Sp1) in the radioresistance of GBM cells. Different human GBM cell lines and tumor-bearing mice were exposed to ionizing radiation (IR). Cell survival was determined by the colony formation assay. The expression of genes and proteins in the cells and tissues was analyzed by RT-PCR and western blotting, respectively. The γ-H2AX, p-Sp1 and dependent protein kinase catalytic subunit (DNA-PKcs phospho S2056) foci were analyzed by immunofluorescence. Apoptotic rates were measured by flow cytometry. Sp1 was upregulated after IR in vitro and in vivo and knocking down Sp1-sensitized GBM cells to IR. Sp1 activated the DNA-PKcs promoter and increased its expression and activity. Furthermore, the loss of Sp1 delayed double-strand breaks (DSB) repair and increased IR-induced apoptosis of GBM cells. Taken together, IR upregulates Sp1 expression in GBM cells, enhancing the activity of DNA-PKcs and promoting IR-induced DSB repair, thereby leading to increased radioresistance.
Collapse
Affiliation(s)
- Xiongxiong Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (X.L.); (T.Z.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (X.L.); (T.Z.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiqi Wang
- College of Life Science, Northwest Normal University, Lanzhou 730030, China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (X.L.); (T.Z.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (X.L.); (T.Z.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (X.L.); (T.Z.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
42
|
Achi J, Achi XW, Veintimilla P, Cueva J. Unusual extraneural metastasis of glioblastoma. Surg Neurol Int 2023; 14:218. [PMID: 37404487 PMCID: PMC10316183 DOI: 10.25259/sni_191_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/10/2023] [Indexed: 07/06/2023] Open
Abstract
Background Glioblastoma (GB) is the most common and aggressive malignant brain tumor in adults. Extracranial metastases are very rare, been described in the lungs, soft tissue, or the intraspinal space. Case Description Through a PubMed-based bibliographic search, the authors reviewed the cases reported in the literature to date, emphasizing the epidemiology and pathophysiology of this rare condition. A clinical case of a 46-year-old man with an initial diagnosis of gliosarcoma, who received complete surgical and adjuvant treatment and later recurred as GB with incidental finding of a lung tumor, whose pathology reported metastasis of the primary, is illustrated. Conclusion Understanding the pathophysiology, it is likely that the incidence of extraneural metastases may continue to increase. Considering improvements in diagnostic techniques that allow early diagnosis, as well as advances in neurosurgical therapy and multimodal management with the aim of improving patient survival, the period in which malignant cells can spread and form extracranial metastases could increase. When screening should be performed to detect metastases in these patients is still not clear. The neuro-oncologists should pay attention to the systematic survey for extraneural metastasis of the GB. Timely detection and early treatment improve overall quality of patients' life.
Collapse
Affiliation(s)
- Jimmy Achi
- Department of Neurosurgery, Hospital Clínica Kennedy, Guayaquil, Ecuador
| | - Xavier Wong Achi
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico City, Mexico
| | - Paula Veintimilla
- Department of Medicine, Universidad Espíritu Santo, Samborondon, Ecuador
| | - Janina Cueva
- Department of General Surgery, Hospital Clínica Kennedy, Guayaquil, Ecuador
| |
Collapse
|
43
|
Liu R, Wu J, Guo H, Yao W, Li S, Lu Y, Jia Y, Liang X, Tang J, Zhang H. Post-translational modifications of histones: Mechanisms, biological functions, and therapeutic targets. MedComm (Beijing) 2023; 4:e292. [PMID: 37220590 PMCID: PMC10200003 DOI: 10.1002/mco2.292] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Histones are DNA-binding basic proteins found in chromosomes. After the histone translation, its amino tail undergoes various modifications, such as methylation, acetylation, phosphorylation, ubiquitination, malonylation, propionylation, butyrylation, crotonylation, and lactylation, which together constitute the "histone code." The relationship between their combination and biological function can be used as an important epigenetic marker. Methylation and demethylation of the same histone residue, acetylation and deacetylation, phosphorylation and dephosphorylation, and even methylation and acetylation between different histone residues cooperate or antagonize with each other, forming a complex network. Histone-modifying enzymes, which cause numerous histone codes, have become a hot topic in the research on cancer therapeutic targets. Therefore, a thorough understanding of the role of histone post-translational modifications (PTMs) in cell life activities is very important for preventing and treating human diseases. In this review, several most thoroughly studied and newly discovered histone PTMs are introduced. Furthermore, we focus on the histone-modifying enzymes with carcinogenic potential, their abnormal modification sites in various tumors, and multiple essential molecular regulation mechanism. Finally, we summarize the missing areas of the current research and point out the direction of future research. We hope to provide a comprehensive understanding and promote further research in this field.
Collapse
Affiliation(s)
- Ruiqi Liu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jiajun Wu
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Haiwei Guo
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Weiping Yao
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Shuang Li
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentJinzhou Medical UniversityJinzhouLiaoningChina
| | - Yanwei Lu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Yongshi Jia
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Xiaodong Liang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jianming Tang
- Department of Radiation OncologyThe First Hospital of Lanzhou UniversityLanzhou UniversityLanzhouGansuChina
| | - Haibo Zhang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| |
Collapse
|
44
|
Wang X, Wang Y, Yu J, Qiu Q, Liao R, Zhang S, Luo C. Reduction-Hypersensitive Podophyllotoxin Prodrug Self-Assembled Nanoparticles for Cancer Treatment. Pharmaceutics 2023; 15:784. [PMID: 36986645 PMCID: PMC10058384 DOI: 10.3390/pharmaceutics15030784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Podophyllotoxin (PPT) has shown strong antitumor effects on various types of cancers. However, the non-specific toxicity and poor solubility severely limits its clinical transformation. In order to overcome the adverse properties of PPT and explore its clinical potential, three novel PTT-fluorene methanol prodrugs linked by different lengths of disulfide bonds were designed and synthesized. Interestingly, the lengths of the disulfide bond affected the drug release, cytotoxicity, pharmacokinetic characteristics, in vivo biodistribution and antitumor efficacy of prodrug NPs. To be more specific, all three PPT prodrugs could self-assemble into uniform nanoparticles (NPs) with high drug loading (>40%) via the one-step nano precipitation method, which not only avoids the use of surfactants and cosurfactants, but also reduces the systemic toxicity of PPT and increases the tolerated dose. Among the three prodrug NPs, FAP NPs containing α-disulfide bond showed the most sensitive tumor-specific response and fastest drug release rate, thus demonstrating the strongest in vitro cytotoxicity. In addition, three prodrug NPs showed prolonged blood circulation and higher tumor accumulation. Finally, FAP NPs demonstrated the strongest in vivo antitumor activity. Our work will advance the pace of podophyllotoxin towards clinical cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
45
|
Alvi MA, Elkaim LM, Levett JJ, Pando A, Roy S, Samuel N, Alotaibi NM, Zadeh G. Current landscape of social media use pertaining to glioblastoma by various stakeholders. Neurooncol Adv 2023; 5:vdad039. [PMID: 37250621 PMCID: PMC10209009 DOI: 10.1093/noajnl/vdad039] [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] [Indexed: 05/31/2023] Open
Abstract
Background Given the potential for social media to allow widespread public engagement, its role in healthcare, including in cancer care as a support network, is garnering interest. To date, the use of social media in neuro-oncology has not been systematically explored. In the current manuscript, we sought to review Twitter use on glioblastoma among patients, caregivers, providers, researchers, and other stakeholders. Methods The Twitter application programming interface (API) database was surveyed from inception to May 2022 to identify tweets about glioblastoma. Number of tweet likes, retweets, quotes, and total engagement were noted for each tweet. Geographic location, number of followers, and number of Tweets were noted for users. We also categorized Tweets based on their underlying themes. A natural language processing (NLP) algorithm was used to assign a polarity score, subjectivity score, and analysis label to each Tweet for sentiment analysis. Results A total of 1690 unique tweets from 1000 accounts were included in our analyses. The frequency of tweets increased from 2013 and peaked in 2018. The most common category among users was MD/researchers (21.6%, n = 216), followed by Media/News (20%, n = 200) and Business (10.7%); patients or caregivers accounted for only 4.7% (n = 47) while medical centers, journals, and foundations accounted for 5.4%, 3.7%, and 2.1%. The most common subjects that Tweets covered included research (54%), followed by personal experience (18.2%) and raising awareness (14%). In terms of sentiment, 43.6% of Tweets were classified as positive, 41.6% as neutral, and 14.9% as negative; a subset analysis of "personal experience" tweets revealed a higher proportion of negative Tweets (31.5%) and less neutral tweets (25%). Only media (β = 8.4; 95% CI [4.4, 12.4]) and follower count (minimally) predicted higher levels of Tweet engagement. Conclusion This comprehensive analysis of tweets on glioblastoma found that the academic community are the most common user group on Twitter. Sentiment analysis revealed that most negative tweets are related to personal experience. These analyses provide the basis for further work into supporting and developing the care of patients with glioblastoma.
Collapse
Affiliation(s)
- Mohammed Ali Alvi
- Divisionof Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Lior M Elkaim
- Corresponding Author: Lior M. Elkaim, MD, Department of Neurology and Neurosurgery, McGill University, 1001 Boulevard Decarie, Montreal, Quebec, Canada ()
| | - Jordan J Levett
- Department of Medicine, University of Montreal, Quebec, Canada
| | - Alejandro Pando
- Department of Neurosurgery, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Sabrina Roy
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Nardin Samuel
- Divisionof Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Naif M Alotaibi
- Department of Neurosurgery, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Gelareh Zadeh
- Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
El-Tanani M, Al Khatib AO, Al-Najjar BO, Shakya AK, El-Tanani Y, Lee YF, Serrano-Aroca Á, Mishra V, Mishra Y, Aljabali AA, Goyal R, Negi P, Farani MR, Binabaj MM, Gholami A, Binabaj MM, Charbe NB, Tambuwala MM. Cellular and molecular basis of therapeutic approaches to breast cancer. Cell Signal 2023; 101:110492. [PMID: 36241056 DOI: 10.1016/j.cellsig.2022.110492] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022]
Abstract
In recent decades, there has been a significant amount of research into breast cancer, with some important breakthroughs in the treatment of both primary and metastatic breast cancers. It's a well-known fact that treating breast cancer is still a challenging endeavour even though physicians have a fantastic toolset of the latest treatment options at their disposal. Due to limitations of current clinical treatment options, traditional chemotherapeutic drugs, and surgical options are still required to address this condition. In recent years, there have been several developments resulting in a wide range of treatment options. This review article discusses the cellular and molecular foundation of chemotherapeutic drugs, endocrine system-based treatments, biological therapies, gene therapy, and innovative techniques for treating breast cancer.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan; Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK.
| | - Arwa Omar Al Khatib
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Belal O Al-Najjar
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Ashok K Shakya
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Yahia El-Tanani
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Yin-Fai Lee
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; School of Life Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Yachana Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 566, Jordan
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Marzieh Ramezani Farani
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), 1417614411 Tehran, Iran.
| | - Maryam Moradi Binabaj
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Amir Gholami
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maryam Moradi Binabaj
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nitin B Charbe
- Center for pharmacometrics and system pharmacology, department of pharmaceutics, college of pharmacy, University of Florida, FL, USA
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK.
| |
Collapse
|
47
|
Xuan Z, Fang L, Zhang G, Zhang X, Jiang J, Wang K, Huang P. The Heterogeneity of Tumour-Associated Macrophages Contributes to the Recurrence and Outcomes of Glioblastoma Patients. J Mol Neurosci 2023; 73:1-14. [PMID: 36542317 DOI: 10.1007/s12031-022-02081-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022]
Abstract
Cellular heterogeneity and immune cell molecular phenotypes may be involved in the malignant progression of glioblastoma (GBM). In this study, we aimed to know whether the heterogeneity of tumour-associated macrophages contributes to the recurrence and outcomes of glioblastoma patients. Single-cell RNA sequencing (scRNA-Seq) data were used to assess the heterogeneity of CD45 + immune cells in recurrent GBM and analyse differentially expressed genes (DEGs) in master cells. Then, a prognostic signature based on the identified DEGs was established and validated, the correlation between risk score and tumour microenvironment (TME) was explored. The correlation between immune infiltration and LGMN, an important DEG in GBM tumour-associated macrophages (TAMs) was illuminated, using integrated bioinformatics analyses. Finally, immunohistochemistry and multiplex immunohistochemistry (mIHC) were used to analyse the expression of LGMN in GBM tissues from our hospital. scRNA-Seq analysis showed that the heterogeneity of recurrent GBM mainly comes from TAMs, which can be divided into 8 cell subclusters. Among these subclusters, TAM1 (markers: CXCL10, ADORA3), TAM3 (markers: MRC1, CFP), TAM4 (markers: GPNMB, PLTP), and TAM5 (markers: CCL4, IRAK2) were specifically present in recurrent GBM. After 342 DEGs in TAMs were identified, a prognostic signature was established based on 13 TAM-associated DEGs, and this signature could serve as an excellent prognostic predictor for patients with GBM. LGMN, one of 13 TAM-associated DEGs, was an important gene in lysosome pathway, we found that macrophage infiltration levels were higher after LGMN upregulation. GBM tissues from our hospital were collected for histopathologic validation, then LGMN was co-expressed with CD68, which is associated with the immune regulation of GBM. In conclusion, cell heterogeneity of TAMs is important for recurrent GBM, a prognostic signature based on 13 TAM-related DEGs can predict the survival outcome of GBM patients. An important DEG, LGMN may regulate the immune cell infiltration of GBM.
Collapse
Affiliation(s)
- Zixue Xuan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Ling Fang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guobing Zhang
- Quality Management Office, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Xin Zhang
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jinying Jiang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| | - Kai Wang
- Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China. .,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| |
Collapse
|
48
|
Belchior A, Fernandes A, Lamotte M, da Silva AFF, Seixas RSGR, Silva AMS, Marques F. Exploring the Physical and Biological Aspects of BNCT with a Carboranylmethylbenzo[ b]acridone Compound in U87 Glioblastoma Cells. Int J Mol Sci 2022; 23:ijms232314929. [PMID: 36499256 PMCID: PMC9737597 DOI: 10.3390/ijms232314929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Boron neutron capture therapy (BNCT) is a re-emerging technique for selectively killing tumor cells. Briefly, the mechanism can be described as follows: after the uptake of boron into cells, the thermal neutrons trigger the fission of the boron atoms, releasing the α-particles and recoiling lithium particles and high-energy photons that damage the cells. We performed a detailed study of the reactor dosimetry, cellular dose assessment, and radiobiological effects induced by BNCT in glioblastoma (GBM) cells. At maximum reactor power, neutron fluence rates were ϕ0 = 6.6 × 107 cm−2 s−1 (thermal) and θ = 2.4 × 104 cm−2 s−1 with a photon dose rate of 150 mGy·h−1. These values agreed with simulations to within 85% (thermal neutrons), 78% (epithermal neutrons), and 95% (photons), thereby validating the MCNPX model. The GEANT4 simulations, based on a realistic cell model and measured boron concentrations, showed that >95% of the dose in cells was due to the BNC reaction. Carboranylmethylbenzo[b]acridone (CMBA) is among the different proposed boron delivery agents that has shown promising properties due to its lower toxicity and important cellular uptake in U87 glioblastoma cells. In particular, the results obtained for CBMA reinforce radiobiological effects demonstrating that damage is mostly induced by the incorporated boron with negligible contribution from the culture medium and adjacent cells, evidencing extranuclear cell radiosensitivity.
Collapse
Affiliation(s)
- Ana Belchior
- Centre for Nuclear Sciences and Technologies, Instituto Superior Técnico, Lisbon University, Nuclear and Technological Campus, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal
| | - Ana Fernandes
- Centre for Nuclear Sciences and Technologies, Instituto Superior Técnico, Lisbon University, Nuclear and Technological Campus, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal
- Department of Nuclear Sciences and Engineering, Instituto Superior Técnico, Lisbon University, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal
| | - Maxime Lamotte
- Centre for Nuclear Sciences and Technologies, Instituto Superior Técnico, Lisbon University, Nuclear and Technological Campus, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal
| | | | | | - Artur M. S. Silva
- Department of Chemistry QOPNA, Aveiro University, 3810-193 Aveiro, Portugal
| | - Fernanda Marques
- Centre for Nuclear Sciences and Technologies, Instituto Superior Técnico, Lisbon University, Nuclear and Technological Campus, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal
- Department of Nuclear Sciences and Engineering, Instituto Superior Técnico, Lisbon University, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal
- Correspondence:
| |
Collapse
|
49
|
Su J, Yao Z, Chen Z, Zhou S, Wang Z, Xia H, Liu S, Wu Y. TfR Aptamer Enhanced Blood-Brain Barrier Penetration of Biomimetic Nanocomplexes for Intracellular Transglutaminase 2 Imaging and Silencing in Glioma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203448. [PMID: 35980938 DOI: 10.1002/smll.202203448] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Engineering a versatile nanocomplex integrating effective penetration of the blood-brain barrier (BBB), accurate diagnosis, and boosting therapy has always been an intractable challenge in glioblastoma multiforme (GBM). Herein, biomimetic nanocomplexes (TMPsM) for single intracellular transglutaminase 2 (TG2)-triggered self-assembly imaging and RNAi therapy for GBM are subtly developed. To prove the concept, transferrin receptor (TfR) aptamer-modified brain metastatic tumor cell membrane is prepared as the shell for dual BBB targeting capability and prolonged blood retention time. Upon targeting entering into GBM, hollow MnO2 is decomposed to release KKGKGQQ-tetraphenylethene (Pep-TPE) and siRNA. Owing to TG2 dependence, the non-emissive Pep-TPE would be self-aggregated to induce the emission turn-on in GBM that contain overexpressed TG2. The resulting aggregation-induced emission fluorescence imaging with a high signal-to-noise ratio can achieve the precise localization of the tumor and dynamic detection of TG2 activity, thereby allowing the GBM accurate diagnosis. Notably, the TG2 can be silenced by the released siRNA to cause cell apoptosis and increase chemotherapeutic sensitivity, ultimately realizing excellent antitumor efficacy. In vitro and in vivo results demonstrate that the as-prepared TMPsM indeed possess superior BBB penetration, precise diagnosis, and effective therapy of GBM. The proposed strategy may pioneer a new path for the theranostics of brain tumors.
Collapse
Affiliation(s)
- Juan Su
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Zhipeng Yao
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- The Translational Research Institute for Neurological Disorders, Department of Neurosurgery of Wannan Medical College, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Zixuan Chen
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Sisi Zhou
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Zhi Wang
- Testing and Certification, Wuxi Institute of Inspection, Wuxi, 214125, China
| | - Hongping Xia
- The Translational Research Institute for Neurological Disorders, Department of Neurosurgery of Wannan Medical College, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Yafeng Wu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| |
Collapse
|
50
|
Guo X, Sui R, Piao H. Tumor-derived small extracellular vesicles: potential roles and mechanism in glioma. J Nanobiotechnology 2022; 20:383. [PMID: 35999601 PMCID: PMC9400220 DOI: 10.1186/s12951-022-01584-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/02/2022] [Indexed: 12/05/2022] Open
Abstract
Small extracellular vesicles (SEVs) are extracellular vesicles containing DNA, RNA, and proteins and are involved in intercellular communication and function, playing an essential role in the growth and metastasis of tumors. SEVs are present in various body fluids and can be isolated and extracted from blood, urine, and cerebrospinal fluid. Under both physiological and pathological conditions, SEVs can be released by some cells, such as immune, stem, and tumor cells, in a cytosolic manner. SEVs secreted by tumor cells are called tumor-derived exosomes (TEXs) because of their origin in the corresponding parent cells. Glioma is the most common intracranial tumor, accounting for approximately half of the primary intracranial tumors, and is characterized by insidious onset, high morbidity, and high mortality rate. Complete removal of tumor tissues by surgery is difficult. Chemotherapy can improve the survival quality of patients to a certain extent; however, gliomas are prone to chemoresistance, which seriously affects the prognosis of patients. In recent years, TEXs have played a vital role in the occurrence, development, associated immune response, chemotherapy resistance, radiation therapy resistance, and metastasis of glioma. This article reviews the role of TEXs in glioma progression, drug resistance, and clinical diagnosis.
Collapse
Affiliation(s)
- Xu Guo
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Shenyang, 110042, Liaoning, China
| | - Rui Sui
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), No. 44 Xiaoheyan Road, Shenyang, 110042, Liaoning, China
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Shenyang, 110042, Liaoning, China.
| |
Collapse
|