1
|
Wang Y, Yu Y, Yu J, Wang C, Wang Y, Fu R, Zhang C. The intersections between neuroscience and medulloblastoma. Cancer Lett 2025; 620:217660. [PMID: 40154912 DOI: 10.1016/j.canlet.2025.217660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Medulloblastoma (MB) represents the most common malignant central nervous system tumor in childhood. The nervous system plays a critical role in the progression of MB, with interactions between the nervous system and cancer significantly influencing oncogenesis, tumor growth, invasion, stemness, and metabolism. These interactions also regulate angiogenesis, metastatic dissemination, the tumor immune microenvironment, and drug resistance. Investigating the nervous system-MB axis holds promise for identifying diagnostic markers, prognostic biomarkers, and therapeutic targets. It also provides insights into the molecular mechanisms underlying MB and informs the development of novel therapeutic strategies. This review summarizes the latest advancements in understanding the interplay between the nervous system and MB, including the role of glial cells in MB and the potential of drug repurposing targeting nervous system components for MB treatment. These findings underscore promising diagnostic and therapeutic opportunities for MB management. Additionally, we outline future research directions in neurosciences that may pave the way for innovative therapeutic approaches and deepen our understanding of this complex disease.
Collapse
Affiliation(s)
- Yafei Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ying Yu
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiahua Yu
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Cheng Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunkun Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Runxi Fu
- Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Institute for Pediatric Research, Shanghai, China
| | - Chenran Zhang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
2
|
Hutson HK, Qin G, Cai C, Nestorova GG. Comparative proteomic profiling of glioblastoma and healthy brain cell-derived extracellular vesicles reveals enrichment of cancer-associated proteins. J Proteomics 2025; 316:105418. [PMID: 40058457 DOI: 10.1016/j.jprot.2025.105418] [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: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs)-mediated cellular communication plays a role in cancer development and progression. This study focuses on identifying glioblastoma-specific EV protein markers through a comparative mass spectrometry bottom-up proteomic analysis of the LN-229 cell line and human neurons, astrocytes, and endothelial brain cells (HEBCs) using timsTOF Pro 2 instrument. The statistically significant upregulated proteins with fold change greater than 2 in the glioblastoma-derived EVs were clustered based on physical and functional interactions using the STRING database and analyzed using Gene Ontology enrichment. LN229-derived EVs contained an average of 2635 proteins, while human astrocytes, neurons, and HEBC encapsulated 2647, 716, and 2285 proteins, respectively. NanoParticle Tracking Analysis indicated that glioblastoma-derived EVs exhibited greater size variability compared to EVs from healthy cells. Statistical analysis identified 25 statistically significant proteins with increased levels in LN229 EVs relative to at least two healthy cell lines suggesting their potential as glioblastoma markers. Functional clustering using the STRING database and GO analysis indicated involvement in epigenetic regulation, metastasis, angiogenesis, and protein folding. Post-translational modification analysis identified a subset of 17 proteins unique to the cancer-derived EVs involved in chromatin regulation, extracellular matrix remodeling, and basement membrane organization pathways, highlighting their role in tumor progression.
Collapse
Affiliation(s)
- Hope K Hutson
- Molecular Science and Nanotechnology, Louisiana Tech University, United States
| | - Guoting Qin
- College of Optometry, University of Houston, United States
| | - Chengzhi Cai
- Department of Chemistry, University of Houston, United States
| | | |
Collapse
|
3
|
Vejzovic D, Kubin A, Fechter K, Karner C, Hartmann J, Ackerbauer T, Radović B, Ritter G, Üçal M, Ropele S, Ali KM, Mischkulnig M, Leoni M, Liegl-Atzwanger B, Wightman L, Rinner B. Glioblastoma targeting by water-soluble hypericin derivate HHL-PVP and photodynamic tumour killing. Biomed Pharmacother 2025; 186:118041. [PMID: 40228364 DOI: 10.1016/j.biopha.2025.118041] [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: 01/07/2025] [Revised: 03/12/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
Hypericin, a tumour-selective photosensitizer, has shown potential in cancer therapy, but its poor water solubility has limited clinical use. To address this, we developed a water-soluble variant called high hypericin-loaded polyvinylpyrrolidone (HHL-PVP) to enhance hypericin's applicability, particularly for treating glioblastoma, a typically terminal disease. We tested HHL-PVP in both in vitro and in vivo models, first confirming its fluorescent properties in the lab and then assessing its efficacy in more complex animal models. Using subcutaneous and orthotopic tumour mouse models, we combined HHL-PVP administration with fluorescence-guided surgery and photodynamic therapy (PDT) to target residual tumour cells. Histological analysis of both healthy and tumour tissue showed HHL-PVP's over 97 % sensitivity and 100 % specificity in distinguishing tumour tissue. In subcutaneous glioblastoma models, significant tumour necrosis and remission occurred after HHL-PVP administration and a 20-minute white light application through the skin. These results highlight HHL-PVP's effectiveness in targeting and eradicating glioblastoma cells. Our findings provide strong evidence that HHL-PVP is a promising therapeutic option for glioblastoma, with its high sensitivity, specificity, and potential for tumour remission through PDT. This approach warrants further investigation in clinical trials and could improve outcomes for a disease that has been difficult to treat.
Collapse
Affiliation(s)
- Djenana Vejzovic
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | | | - Karoline Fechter
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | - Christina Karner
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | | | | | - Branislav Radović
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gerald Ritter
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | - Muammer Üçal
- Department of Neurosurgery, Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria; Department of Neurology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Kariem Mahdy Ali
- Department of Neurosurgery, Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | | | - Marlene Leoni
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | | | - Beate Rinner
- Division of Biomedical Research, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria.
| |
Collapse
|
4
|
Tang Z, Xue Z, Liu X, Zhang Y, Zhao J, Liu J, Zhang L, Guo Q, Feng B, Wang J, Zhang D, Li X. Inhibition of hypoxic exosomal miR-423-3p decreases glioma progression by restricting autophagy in astrocytes. Cell Death Dis 2025; 16:265. [PMID: 40199864 PMCID: PMC11978802 DOI: 10.1038/s41419-025-07576-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025]
Abstract
The tumor microenvironment (TME) of gliomas comprises glioma cells and surrounding cells, such as astrocytes, macrophages, T cells, and neurons. In the TME, glioma cells can activate normal human astrocytes (NHAs) through the secretion of exosomes and the activation of astrocytes can further improve the progression of glioma, leading to a poor prognosis for patients. However, the molecular mechanisms underlying NHAs activation by gliomas remain largely unknown. It this study, glioma-derived exosomes (GDEs) play an important role in the modulation of autophagy and activation of NHAs. Compared with normoxic GDEs, hypoxic glioma-derived exosomes (H-GDEs) further improved autophagy and activation of astrocytes, which strongly promoted the progression of glioma cells. In an miRNA array between two types of exosomes from gliomas, miR-423-3p was highly expressed in H-GDEs and played an important role in autophagy, resulting in the activation of NHAs. The mechanism by which hypoxic glioma cells react with NHAs to create an immunosuppressive microenvironment was identified and 15d-PGJ2 was established as an effective inhibitor of miR-423-3p to suppress NHAs activation. These findings provide new insights into the diagnosis and treatment of gliomas by targeting autophagy and miR-423-3p expression.
Collapse
Affiliation(s)
- Ziyi Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Xuchen Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiangli Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Junzhi Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Lin Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qindong Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Bowen Feng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| |
Collapse
|
5
|
Wang C, Fan X, Shi Y, Tang F. Radiation-Induced Brain Injury with Special Reference to Astrocytes as a Therapeutic Target. J Integr Neurosci 2025; 24:25907. [PMID: 40152565 DOI: 10.31083/jin25907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 03/29/2025] Open
Abstract
Radiotherapy is one of the primary modalities for oncologic treatment and has been utilized at least once in over half of newly diagnosed cancer patients. Cranial radiotherapy has significantly enhanced the long-term survival rates of patients with brain tumors. However, radiation-induced brain injury, particularly hippocampal neuronal damage along with impairment of neurogenesis, inflammation, and gliosis, adversely affects the quality of life for these patients. Astrocytes, a type of glial cell that are abundant in the brain, play essential roles in maintaining brain homeostasis and function. Despite their importance, the pathophysiological changes in astrocytes induced by radiation have not been thoroughly investigated, and no systematic or comprehensive review addressing the effects of radiation on astrocytes and related diseases has been conducted. In this paper, we review current studies on the neurophysiological roles of astrocytes following radiation exposure. We describe the pathophysiological changes in astrocytes, including astrogliosis, astrosenescence, and the associated cellular and molecular mechanisms. Additionally, we summarize the roles of astrocytes in radiation-induced impairments of neurogenesis and the blood-brain barrier (BBB). Based on current research, we propose that brain astrocytes may serve as potential therapeutic targets for treating radiation-induced brain injury (RIBI) and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Caiping Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Xingjuan Fan
- Department of Neurology, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, China
| | - Yunwei Shi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| |
Collapse
|
6
|
Kiran S, Xue Y, Sarker DB, Sang QXA. Effects of Induced Pluripotent Stem Cell-Derived Astrocytes on Cisplatin Sensitivity in Pediatric Brain Cancer Cells. Cancers (Basel) 2025; 17:997. [PMID: 40149331 PMCID: PMC11940393 DOI: 10.3390/cancers17060997] [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: 01/27/2025] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background: ATRTs and DIPGs are deadly pediatric brain tumors with poor prognosis. These tumors can develop resistance to chemotherapies, which may be significantly influenced by their microenvironment. Since astrocytes are the most abundant glial cell type in the brain microenvironment and may support tumor growth and chemoresistance, this study investigated the effects of induced pluripotent stem cell-derived astrocytes (iPSC-astrocytes) on cisplatin sensitivity in CHLA-05-ATRT and SF8628 (DIPG) cells. iPSCs provide an unlimited and standardized source of nascent astrocytes, which enables modeling the interaction between childhood brain tumor cells and iPSC-astrocytes within a controlled coculture system. Methods: To study the effects on tumor growth, the iPSC-astrocytes were cocultured with tumor cells. Additionally, the tumor cells were exposed to various concentrations of cisplatin to evaluate their chemosensitivity in the presence of astrocytes. Results: The paracrine interaction of iPSC-astrocytes with tumor cells upregulated astrocyte activation markers GFAP and STAT3 and promoted tumor cell proliferation. Moreover, the cisplatin treatment significantly decreased the viability of CHLA-05-ATRT and SF8628 cells. However, tumor cells exhibited reduced sensitivity to cisplatin in the coculture with iPSC-astrocytes. During cisplatin treatment, DIPG cells in particular showed upregulation of resistance markers, ERK1, STAT3, and MTDH, which are associated with enhanced proliferation and invasion. They also had increased expression of APEX1, which is involved in the base excision repair pathway following cisplatin-induced DNA damage. Conclusion: These findings underscore the significance of the tumor microenvironment in modulating tumor cell survival and chemosensitivity.
Collapse
Affiliation(s)
- Sonia Kiran
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
7
|
Gui Y, Qin H, Zhang X, Chen Q, Ye F, Tian G, Yang S, Ye Y, Pan D, Zhou J, Fan X, Wang Y, Zhao L. Glioma-astrocyte connexin43 confers temozolomide resistance through activation of the E2F1/ERCC1 axis. Neuro Oncol 2025; 27:711-726. [PMID: 39514365 PMCID: PMC11889727 DOI: 10.1093/neuonc/noae237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Glioma is the most prevalent and lethal tumor of the central nervous system. Routine treatment with temozolomide (TMZ) would unfortunately result in inevitable recurrence and therapy resistance, severely limiting therapeutic efficacy. Tumor-associated astrocytes (TAAs) are key components of the tumor microenvironment and increasing evidence has demonstrated that aberrant expression of connexin43 (Cx43) was closely associated with glioma progression and TMZ resistance. However, the specific role of Cx43 in mediating TMZ resistance through glioma and astrocyte interactions has not been fully explored. METHODS The expression and prognostic value of Cx43 were evaluated in tumor samples and clinical databases. ShRNA-medicated knockdown and Gfap-Cre Cx43flox/flox gene mouse were used to assess the role and functional significance of Cx43 in vitro and in vivo. Moreover, we performed mass spectrometry analysis, chromatin immunoprecipitation, and other biochemical assays to define the molecular mechanisms by which Cx43 promotes TMZ resistance. RESULTS We confirmed that the upregulation of Cx43 expression between TAAs and glioma cells contributed to TMZ resistance and tumor recurrence. Genetic knockdown or pharmacological inhibition of Cx43 enhanced TMZ-induced cytotoxicity. Mechanistically, elevated Cx43 expression induced β-catenin accumulation at the cell surface of glioma cells, suppressing T-cell factor/lymphoid enhancer-binding factor transcription. This led to impaired miR-205-5p expression and subsequent activation of the E2F1/ERCC1 axis, which eventually led to chemoresistance. CONCLUSIONS Our study reveals a novel regulatory mechanism in which the Cx43/miR-205-5p/E2F1/ERCC1 axis contributes to TMZ resistance in glioma. These findings further highlight the potential of targeting Cx43 as a therapeutic strategy in glioma.
Collapse
Affiliation(s)
- Yanping Gui
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Hongkun Qin
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Xinyu Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Qianqian Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Fangyu Ye
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Geng Tian
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Shihe Yang
- Public Experimental Platform, China Pharmaceutical University, Nanjing, P.R. China
| | - Yuting Ye
- Public Experimental Platform, China Pharmaceutical University, Nanjing, P.R. China
| | - Di Pan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, P.R. China
| | - Jieying Zhou
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida
| | - Xiangshan Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, P.R. China
| | - Yajing Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Li Zhao
- Public Experimental Platform, China Pharmaceutical University, Nanjing, P.R. China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
8
|
López de Mingo I, Rivera González MX, Ramos Gómez M, Maestú Unturbe C. The Frequency of a Magnetic Field Determines the Behavior of Tumor and Non-Tumor Nerve Cell Models. Int J Mol Sci 2025; 26:2032. [PMID: 40076656 PMCID: PMC11899782 DOI: 10.3390/ijms26052032] [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: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The involvement of magnetic fields in basic cellular processes has been studied for years. Most studies focus their results on a single frequency and intensity. Intensity has long been the central parameter in hypotheses of interaction between cells and magnetic fields; however, frequency has always played a secondary role. The main objective of this study was to obtain a specific frequency that allows a reduction in the viability and proliferation of glioblastoma (CT2A) and neuroblastoma (N2A) cell models. These were compared with an astrocyte cell model (C8D1A) (nontumor) to determine whether there is a specific frequency of response for each of the cell lines used. The CT2A, C8D1A, and N2A cell lines were exposed to a magnetic field of 100 µT and a variable frequency range between 20 and 100 Hz for 24, 48 and 72 h. The results fit a biological window model in which the viability and proliferation of N2A and CT2A cells decrease statistically significantly in a 50 Hz center of value window. In addition, the non-tumor cell model showed different behavior from tumor cell models depending on the applied frequency. These results are promising in the use of magnetic fields for therapeutic purposes.
Collapse
Affiliation(s)
- Isabel López de Mingo
- Escuela Técnica Superior de Ingenieros de Telecomunicación (ETSIT), Universidad Politécnica de Madrid, 28040 Madrid, Spain; (I.L.d.M.); (M.R.G.)
- Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain;
| | - Marco-Xavier Rivera González
- Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain;
- Escuela Técnica Superior de Ingenieros Informáticos (ETSIINF), Universidad Politécnica de Madrid, 28223 Madrid, Spain
| | - Milagros Ramos Gómez
- Escuela Técnica Superior de Ingenieros de Telecomunicación (ETSIT), Universidad Politécnica de Madrid, 28040 Madrid, Spain; (I.L.d.M.); (M.R.G.)
- Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain;
- Centro de Investigación Biomédica en Red (CIBER-BBN), 28029 Madrid, Spain
| | - Ceferino Maestú Unturbe
- Escuela Técnica Superior de Ingenieros de Telecomunicación (ETSIT), Universidad Politécnica de Madrid, 28040 Madrid, Spain; (I.L.d.M.); (M.R.G.)
- Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain;
- Centro de Investigación Biomédica en Red (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
9
|
He Q, Hu J, Ngo FY, Zhang H, He L, Huang H, Wu T, Pan Y, Yang Z, Jiang Y, Cho WC, Cheuk W, Tse GM, Tsang JY, Yang M, Zhang L, Wang X, Lo PC, Lau CG, Chin YR. Targeting TUBB2B inhibits triple-negative breast cancer growth and brain-metastatic colonization. J Exp Clin Cancer Res 2025; 44:55. [PMID: 39962586 PMCID: PMC11831766 DOI: 10.1186/s13046-025-03312-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 02/01/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The triple-negative subtype of breast cancer is particularly challenging to treat due to its aggressiveness with a high risk of brain metastasis, and the lack of effective targeted therapies. Tubulin beta 2B class IIb (TUBB2B), a β-tubulin isoform regulating axon guidance during embryonic development, was found to be overexpressed in various types of cancers including triple-negative breast cancer (TNBC). However, its functional roles in breast cancer or metastasis remain unclear. METHODS To identify TUBB2B as a novel molecular target in TNBC, we performed bioinformatics analysis to assess the association of TUBB2B expression and survival of patients. RNAscope in situ hybridization was used to examine TUBB2B expression in clinical breast tumor samples. The effect of TUBB2B knockdown on TNBC growth and brain metastasis colonization was evaluated by in vitro and in vivo assays. Mass spectrometry (MS) and biochemical experiments were performed to explore the underlying mechanisms. Preclinical efficacy of targeting TUBB2B was determined in xenograft studies using the siRNA-gold nanoparticle (siRNA-AuNP) approach. RESULTS TUBB2B, but not other β-tubulin isoforms, is frequently overexpressed in TNBC primary tumors as well as brain metastases. We also find that upregulation of TUBB2B is associated with poor prognosis in breast cancer patients. Silencing TUBB2B induces tumor cell death and inhibits the outgrowth of brain metastasis. Mechanistically, we identify eukaryotic translation elongation factor 1 alpha 1 (eEF1A1) as a novel interacting partner of TUBB2B, revealing a previously unexplored role of TUBB2B in translational regulation. In line with its neural-related functions, TUBB2B overexpression in TNBC cells activates astrocytes, which in turn upregulate TUBB2B in tumor cells. These findings suggest a feed-forward interaction between TUBB2B in TNBC cells and astrocytes that promotes brain metastatic colonization. Furthermore, we demonstrate the potent inhibition of TNBC xenograft growth as well as brain metastatic colonization using TUBB2B siRNA-AuNP treatment, indicating potential clinical applications of targeting TUBB2B for TNBC. CONCLUSIONS TUBB2B is a novel TNBC gene that plays a key role in promoting tumor cell survival and brain metastatic colonization, and can be targeted by siRNA-AuNPs as a treatment strategy.
Collapse
Affiliation(s)
- Qingling He
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China
| | - Jianyang Hu
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China
| | - Fung-Yin Ngo
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Huiqi Zhang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Lin He
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Hao Huang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Tan Wu
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Yilin Pan
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Zihan Yang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Yuanyuan Jiang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon Tong, Hong Kong
| | - Wah Cheuk
- Department of Pathology, Queen Elizabeth Hospital, Kowloon Tong, Hong Kong
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Julia Y Tsang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Mengsu Yang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Liang Zhang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Sha Tin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Pui-Chi Lo
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China
| | - C Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Y Rebecca Chin
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon Tong, Hong Kong.
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, Guangdong, China.
| |
Collapse
|
10
|
Xu X, Wang J, Chen T, Wang S, Wang F, He J, Meng XY, Shen Y. Deciphering novel mitochondrial signatures: multi-omics analysis uncovers cross-disease markers and oligodendrocyte pathways in Alzheimer's disease and glioblastoma. Front Aging Neurosci 2025; 17:1536142. [PMID: 40018519 PMCID: PMC11865232 DOI: 10.3389/fnagi.2025.1536142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Alzheimer's disease (AD) and glioblastoma (GBM) are severe neurological disorders that pose significant global healthcare challenges. Despite extensive research, the molecular mechanisms, particularly those involving mitochondrial dysfunction, remain poorly understood. A major limitation in current studies is the lack of cell-specific markers that effectively represent mitochondrial dynamics in AD and GBM. Methods In this study, we analyzed single-cell transcriptomic data using 10 machine learning algorithms to identify mitochondria-associated cell-specific markers. We validated these markers through the integration of gene expression and methylation data across diverse cell types. Our dataset comprised single-nucleus RNA sequencing (snRNA-seq) from AD patients, single-cell RNA sequencing (scRNA-seq) from GBM patients, and additional DNA methylation and transcriptomic data from the ROSMAP, ADNI, TCGA, and CGGA cohorts. Results Our analysis identified four significant cross-disease mitochondrial markers: EFHD1, SASH1, FAM110B, and SLC25A18. These markers showed both shared and unique expression profiles in AD and GBM, suggesting a common mitochondrial mechanism contributing to both diseases. Additionally, oligodendrocytes and their interactions with astrocytes were implicated in disease progression, particularly through the APP signaling pathway. Key hub genes, such as HS6ST3 and TUBB2B, were identified across different cellular subpopulations, highlighting a cell-specific co-expression network linked to mitochondrial function.
Collapse
Affiliation(s)
- Xuan Xu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jiaqi Wang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Tong Chen
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui, China
| | - Shuaibin Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui, China
| | - Fei Wang
- School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| | - Junwen He
- College of Informatics, Huazhong Agricultural University, Hubei, Wuhan, China
| | - Xiang-Yu Meng
- School of Basic Medical Sciences, Medical School, Hubei Minzu University, Enshi, Hubei, China
| | - Yin Shen
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
11
|
Franco MS, Raulefs S, Schilling D, Combs SE, Schmid TE. Impact of Radiation on Invasion and Migration of Glioma In Vitro and In Vivo. Cancers (Basel) 2024; 16:3900. [PMID: 39682088 DOI: 10.3390/cancers16233900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Glioblastoma (GBM) constitutes the most common primary brain tumor and it remains incurable despite therapeutic advances. The high infiltration/invasion potential of GBM cells is considered to be one of the reasons for the inevitable recurrence of the disease. Radiotherapy (RT) is part of the standard care for patients with GBM, and its benefits on overall survival are extensively reported. However, numerous preclinical studies show that X-ray irradiation can enhance the motility of GBM cells. In the present review, we bring together state-of-the-art research on the impact of radiation on GBM cell motility. The mechanisms through which irradiation impacts the brain tumor microenvironment and the tumor cells themselves, leading to more aggressive/invasive tumors, are described. Finally, we summarize potential pharmacological strategies to overcome this problem. Clinical data validating the occurrence of these processes are urgently needed as they could be of great value for patient outcomes. With this comprehensive review, we expect to highlight the need for methods which allow for monitoring the post-irradiation invasive behavior of GBM in patients.
Collapse
Affiliation(s)
- Marina Santiago Franco
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Susanne Raulefs
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Daniela Schilling
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Stephanie E Combs
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Thomas E Schmid
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| |
Collapse
|
12
|
Wang M, Graner AN, Knowles B, McRae C, Fringuello A, Paucek P, Gavrilovic M, Redwine M, Hanson C, Coughlan C, Grimaldo-Garcia S, Metzger B, Bolus V, Kopper TJ, Smith M, Zhou W, Lenz M, Abosch A, Ojemann S, Lillehei KO, Yu X, Graner MW. Differential Effects of Extracellular Vesicles from Two Different Glioblastomas on Normal Human Brain Cells. Neurol Int 2024; 16:1355-1384. [PMID: 39585062 PMCID: PMC11587087 DOI: 10.3390/neurolint16060103] [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: 09/05/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Background/Objectives: Glioblastomas (GBMs) are dreadful brain tumors with abysmal survival outcomes. GBM extracellular vesicles (EVs) dramatically affect normal brain cells (largely astrocytes) constituting the tumor microenvironment (TME). We asked if EVs from different GBM patient-derived spheroid lines would differentially alter recipient brain cell phenotypes. This turned out to be the case, with the net outcome of treatment with GBM EVs nonetheless converging on increased tumorigenicity. Methods: GBM spheroids and brain slices were derived from neurosurgical patient tissues following informed consent. Astrocytes were commercially obtained. EVs were isolated from conditioned culture media by ultrafiltration, concentration, and ultracentrifugation. EVs were characterized by nanoparticle tracking analysis, electron microscopy, biochemical markers, and proteomics. Astrocytes/brain tissues were treated with GBM EVs before downstream analyses. Results: EVs from different GBMs induced brain cells to alter secretomes with pro-inflammatory or TME-modifying (proteolytic) effects. Astrocyte responses ranged from anti-viral gene/protein expression and cytokine release to altered extracellular signal-regulated protein kinase (ERK1/2) signaling pathways, and conditioned media from EV-treated cells increased GBM cell proliferation. Conclusions: Astrocytes/brain slices treated with different GBM EVs underwent non-identical changes in various omics readouts and other assays, indicating "personalized" tumor-specific GBM EV effects on the TME. This raises concern regarding reliance on "model" systems as a sole basis for translational direction. Nonetheless, net downstream impacts from differential cellular and TME effects still led to increased tumorigenic capacities for the different GBMs.
Collapse
Affiliation(s)
- Mary Wang
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Arin N. Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Bryne Knowles
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Charlotte McRae
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Anthony Fringuello
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Department of Cell Biology, State University of New York Downstate Health Sciences University, New York, NY 11203, USA
| | - Petr Paucek
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Michael Gavrilovic
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Department of Biomedical Sciences, Regis University, Denver, CO 80221, USA
- St Louis University School of Medicine, St. Louis, MO 63104, USA
| | - McKenna Redwine
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Department of Biomedical Sciences, Regis University, Denver, CO 80221, USA
| | - Caleb Hanson
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Department of Biomedical Sciences, Regis University, Denver, CO 80221, USA
| | - Christina Coughlan
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Stacey Grimaldo-Garcia
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Department of Neuroscience, Middlebury College, Middlebury, VT 05753, USA
| | - Brooke Metzger
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Occupational Therapy, Illinois College, Jacksonville, IL 62650, USA
- Neuroscience, Midwestern University, Glendale, AZ 85308, USA
| | - Vince Bolus
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Timothy J. Kopper
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Marie Smith
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Wenbo Zhou
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Morgan Lenz
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
- Occupational Therapy, Illinois College, Jacksonville, IL 62650, USA
| | - Aviva Abosch
- Department of Neurosurgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Steven Ojemann
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Kevin O. Lillehei
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Xiaoli Yu
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| | - Michael W. Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.W.); (A.N.G.); (B.K.); (C.M.); (A.F.); (P.P.); (M.G.); (M.R.); (C.H.); (S.G.-G.); (B.M.); (V.B.); (T.J.K.); (M.S.); (W.Z.); (M.L.); (S.O.); (K.O.L.); (X.Y.)
| |
Collapse
|
13
|
DePalma TJ, Hisey CL, Hughes K, Fraas D, Tawfik M, Scharenberg J, Wiggins S, Nguyen KT, Hansford DJ, Reátegui E, Skardal A. Tuning a bioengineered hydrogel for studying astrocyte reactivity in glioblastoma. Acta Biomater 2024; 189:155-167. [PMID: 39370091 PMCID: PMC11801334 DOI: 10.1016/j.actbio.2024.09.048] [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: 05/23/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
Astrocytes play many essential roles in the central nervous system (CNS) and are altered significantly in disease. These reactive astrocytes contribute to neuroinflammation and disease progression in many pathologies, including glioblastoma (GB), an aggressive form of brain cancer. Current in vitro platforms do not allow for accurate modeling of reactive astrocytes. In this study, we sought to engineer a simple bioengineered hydrogel platform that would support the growth of primary human astrocytes and allow for accurate analysis of various reactive states. After validating this platform using morphological analysis and qPCR, we then used the platform to begin investigating how astrocytes respond to GB derived extracellular vesicles (EVs) and soluble factors (SF). These studies reveal that EVs and SFs induce distinct astrocytic states. In future studies, this platform can be used to study how astrocytes transform the tumor microenvironment in GB and other diseases of the CNS. STATEMENT OF SIGNIFICANCE: Recent work has shown that astrocytes help maintain brain homeostasis and may contribute to disease progression in diseases such as glioblastoma (GB), a deadly primary brain cancer. In vitro models allow researchers to study basic mechanisms of astrocyte biology in healthy and diseased conditions, however current in vitro systems do not accurately mimic the native brain microenvironment. In this study, we show that our hydrogel system supports primary human astrocyte culture with an accurate phenotype and allows us to study how astrocytes change in response to a variety of inflammatory signals in GB. This platform could be used further investigate astrocyte behavior and possible therapeutics that target reactive astrocytes in GB and other brain diseases.
Collapse
Affiliation(s)
- Thomas J DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Colin L Hisey
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Kennedy Hughes
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - David Fraas
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Marie Tawfik
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jason Scharenberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Sydney Wiggins
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Kim Truc Nguyen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Derek J Hansford
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Eduardo Reátegui
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
14
|
Zhang L, Wang Y, Cai X, Mao X, Sun H. Deciphering the CNS-glioma dialogue: Advanced insights into CNS-glioma communication pathways and their therapeutic potential. J Cent Nerv Syst Dis 2024; 16:11795735241292188. [PMID: 39493257 PMCID: PMC11528668 DOI: 10.1177/11795735241292188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024] Open
Abstract
The field of cancer neuroscience has rapidly evolved, shedding light on the complex interplay between the nervous system and cancer, with a particular focus on the relationship between the central nervous system (CNS) and gliomas. Recent advancements have underscored the critical influence of CNS activity on glioma progression, emphasizing the roles of neurons and neuroglial cells in both the onset and evolution of gliomas. This review meticulously explores the primary communication pathways between the CNS and gliomas, encompassing neuro-glioma synapses, paracrine mechanisms, extracellular vesicles, tunneling nanotubes, and the integrative CNS-immune-glioma axis. It also evaluates current and emerging therapeutic interventions aimed at these pathways and proposes forward-looking perspectives for research in this domain.
Collapse
Affiliation(s)
- Lu Zhang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yajing Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxi Cai
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyuan Mao
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong–Hong Kong–Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Knudsen AM, Ewald JD, Pedersen V, Haupt-Jorgensen M, Hansen EVR, Kristensen BW. Characterisation of the tumour microenvironment in primary and recurrent glioblastomas. Neuropathol Appl Neurobiol 2024; 50:e13012. [PMID: 39449228 DOI: 10.1111/nan.13012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024]
Abstract
AIMS Glioblastoma patients have a dismal prognosis, due to inevitable tumour recurrence and respond poorly to immunotherapy. Tumour-associated microglia/macrophages (TAMs) dominate the glioblastoma tumour microenvironment and have been implicated in tumour progression and immune evasion. Early recurrent glioblastomas contain focal reactive regions with occasional fibrosis, chronic inflammation, TAMs and tumour cells. Surgical specimens from these tumours are rare and provide crucial insights into glioblastoma recurrence biology. This study aimed to characterise TAM- and lymphocyte phenotypes in primary vs early- and late-recurrent glioblastomas. METHODS Patient-matched primary and recurrent glioblastomas were compared between patients with early recurrences (n = 11, recurrence ≤6 months) and late recurrences (n = 12, recurrence after 12-19 months). Double-immunofluorescence stains combining Iba1 with HLA-DR, CD14, CD68, CD74, CD86, CD163, CD204 and CD206 along with stains for CD20, CD3, CD8 and FOXP3 were quantified with software-based classifiers. RESULTS Reactive regions in early recurrent tumours contained more TAMs (31.4% vs 21.7%, P = 0.01), which showed increased expression of CD86 (59.4% vs 38.4%, P = 0.04), CD204 (48.5% vs 28.4%, P = 0.03), CD206 (25.5% vs 14.4%, P = 0.04) and increased staining intensity for CD163 (86.4 vs 57.7 arbitrary units, P = 0.02), compared to late recurring tumours. Reactive regions contained more B-lymphocytes compared to patient-matched primary tumours (0.71% vs 0.40%, P = 0.04). Fractions of total, cytotoxic and regulatory T-lymphocytes did not differ. CONCLUSIONS Early recurrent glioblastomas showed enrichment for TAMs, expressing both pro- and anti-inflammatory markers and B-lymphocytes. This may indicate a time-dependent response to immunotherapy explained by time-dependent alterations in the immune-microenvironment in recurrent glioblastomas.
Collapse
Affiliation(s)
- Arnon Møldrup Knudsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper Dupont Ewald
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Vilde Pedersen
- Department of Pathology, Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine and Biotech Research Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Martin Haupt-Jorgensen
- Department of Pathology, Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Bjarne Winther Kristensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Pathology, Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine and Biotech Research Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Quan W, Xu CS, Ma C, Chen X, Yu DH, Li ZY, Wang DW, Tang F, Wan GP, Wan J, Wang ZF, Li ZQ. Anti-tumor effects of telmisartan in glioma-astrocyte non-contact co-cultures: A critical role of astrocytic IL-6-mediated paracrine growth promotion. Int Immunopharmacol 2024; 139:112707. [PMID: 39032472 DOI: 10.1016/j.intimp.2024.112707] [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: 06/22/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Telmisartan, an angiotensin II type 1 receptor (AT1R) blocker, exhibits broad anti-tumor activity. However, in vitro, anti-proliferative effects are shown at doses far beyond the therapeutic plasma concentration. Considering the role of tumor microenvironment in glioma progression, glioma-astrocyte co-cultures were employed to test the anti-tumor potential of low-dose telmisartan. When a high dose was required for a direct anti-proliferative effect on glioma cell lines, a low dose significantly inhibited glioma cell proliferation and migration in the co-culture system. Under co-culture conditions, upregulated IL-6 expression in astrocytes played a critical role in glioma progression. Silencing IL-6 in astrocytes or IL-6R in glioma cells reduced proliferation and migration. Telmisartan (5 μM) inhibited astrocytic IL-6 expression, and its anti-tumor effects were reversed by silencing IL-6 or IL-6R and inhibiting signal transducer and activator of transcription 3 (STAT3) activity in glioma cells. Moreover, the telmisartan-driven IL-6 downregulation was not imitated by losartan, an AT1R blocker with little capacity of peroxisome proliferator-activated receptor-gamma (PPARγ) activation, but was eliminated by a PPARγ antagonist, indicating that the anti-glioma effects of telmisartan rely on its PPARγ agonistic activity rather than AT1R blockade. This study highlights the importance of astrocytic IL-6-mediated paracrine signaling in glioma growth and the potential of telmisartan as an adjuvant therapy for patients with glioma, especially those with hypertension.
Collapse
Affiliation(s)
- Wei Quan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng-Shi Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xi Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dong-Hu Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhi-Yu Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dan-Wen Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Feng Tang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Gui-Ping Wan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Hubei, China.
| |
Collapse
|
17
|
Xiao T, Yu X, Tao J, Yang L, Duan X. Metabolomics-Based Study of the Protective Effect of 4-Hydroxybenzyl Alcohol on Ischemic Astrocytes. Int J Mol Sci 2024; 25:9907. [PMID: 39337395 PMCID: PMC11432256 DOI: 10.3390/ijms25189907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Ischemic stroke is a common and dangerous disease in clinical practice. Astrocytes (ASs) are essential for maintaining the metabolic balance of the affected regions during the disease process. 4-Hydroxybenzyl alcohol (4HBA) from Gastrodia elata Bl. has potential neuroprotective properties due to its ability to cross the blood-brain barrier. In an in vitro experiment, we replicated the oxygen-glucose deprivation/reoxygenation model, and used methyl thiazoly tertrazolium, flow cytometry, kits, and other technical means to clarify the protective effect of 4HBA on primary ASs. In in vivo experiments, the 2VO model was replicated, and immunofluorescence and immunohistochemistry techniques were used to clarify the protective effect of 4HBA on ASs and the maintenance of the blood-brain barrier. Differential metabolites and related pathways were screened and verified using metabolomics analysis and western blot. 4HBA noticeably amplified AS cell survival, reduced mitochondrial dysfunction, and mitigated oxidative stress. It demonstrated a protective effect on ASs in both environments and was instrumental in stabilizing the blood-brain barrier. Metabolomic data indicated that 4HBA regulated nucleic acid and glutathione metabolism, influencing purines, pyrimidines, and amino acids, and it activated the N-methyl-D-aspartate/p-cAMP-response element binding protein/brain-derived neurotrophic factor signaling pathway via N-methyl-D-aspartate R1/N-methyl-D-aspartate 2C receptors. Our findings suggest that 4HBA is a potent neuroprotective agent against ischemic stroke, enhancing AS cell survival and function while stabilizing the blood-brain barrier. The N-methyl-D-aspartate/p-cAMP-response element binding protein/brain-derived neurotrophic factor signaling pathway is activated by 4HBA.
Collapse
Affiliation(s)
- Tian Xiao
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xingzhi Yu
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Jie Tao
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Liping Yang
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| |
Collapse
|
18
|
Ribeiro JH, Villarinho NJ, Fernandes PV, Spohr TCLDSE, Lopes GPDF. Conditioned Medium From Reactive Astrocytes Inhibits Proliferation, Resistance, and Migration of p53-Mutant Glioblastoma Spheroid Through GLI-1 Downregulation. J Cell Biochem 2024; 125:e30637. [PMID: 39150066 DOI: 10.1002/jcb.30637] [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: 05/13/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
Glioblastoma (GBM) aggressiveness is partly driven by the reactivation of signaling pathways such as Sonic hedgehog (SHH) and the interaction with its microenvironment. SHH pathway activation is one of the phenomena behind the glial transformation in response to tumor growth. The reactivation of the SHH signaling cascade during GBM-astrocyte interaction is highly relevant to understanding the mechanisms used by the tumor to modulate the adjacent stroma. The role of reactive astrocytes considering SHH signaling during GBM progression is investigated using a 3D in vitro model. T98G GBM spheroids displayed significant downregulation of SHH (61.4 ± 9.3%), GLI-1 (6.5 ± 3.7%), Ki-67 (33.7 ± 8.1%), and mutant MTp53 (21.3 ± 10.6%) compared to the CONTROL group when incubated with conditioned medium of reactive astrocytes (CM-AST). The SHH pathway inhibitor, GANT-61, significantly reduced previous markers (SHH = 43.0 ± 12.1%; GLI-1 = 9.5 ± 3.4%; Ki-67 = 31.9 ± 4.6%; MTp53 = 6.5 ± 7.5%) compared to the CONTROL, and a synergistic effect could be observed between GANT-61 and CM-AST. The volume (2.0 ± 0.2 × 107 µm³), cell viability (80.4 ± 3.2%), and migration (41 ± 10%) of GBM spheroids were significantly reduced in the presence of GANT-61 and CM-AST when compared to CM-AST after 72 h (volume = 2.3 ± 0.4 × 107 µm³; viability = 92.2 ± 6.5%; migration = 102.5 ± 14.6%). Results demonstrated that factors released by reactive astrocytes promoted a neuroprotective effect preventing GBM progression using a 3D in vitro model potentiated by SHH pathway inhibition.
Collapse
Affiliation(s)
- Jessica Honorato Ribeiro
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK-CEN, Mol, Antwerp, Belgium
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Nícolas Jones Villarinho
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratory of Tumor Microenvironment, Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), University of São Paulo, São Paulo, Brazil
| | - Priscila Valverde Fernandes
- Department of Pathology, Pathology Division, Instituto Nacional do Câncer (DIPAT-INCA), Rio de Janeiro, Brazil
| | - Tania Cristina Leite de Sampaio E Spohr
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Director of Sample Preparation, Cell Culture and Biobanking, Centogene, Rostock, Germany
| | - Giselle Pinto de Faria Lopes
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Department of Marine Biotechnology, Natural Products Division, Instituto de Estudos do Mar Almirante Paulo Moreira (IEAPM), Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Malone K, Dugas M, Earl N, Alain T, LaCasse EC, Beug ST. Astrocytes and the tumor microenvironment inflammatory state dictate the killing of glioblastoma cells by Smac mimetic compounds. Cell Death Dis 2024; 15:592. [PMID: 39147758 PMCID: PMC11327263 DOI: 10.1038/s41419-024-06971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Smac mimetic compounds (SMCs) are small molecule drugs that sensitize cancer cells to TNF-α-induced cell death and have multiple immunostimulatory effects through alterations in NF-κB signaling. The combination of SMCs with immunotherapies has been reported to result in durable cures of up to 40% in syngeneic, orthotopic murine glioblastoma (GBM) models. Herein, we find that SMC resistance is not due to a cell-intrinsic mechanism of resistance. We thus evaluated the contribution of GBM and brain stromal components to identify parameters leading to SMC efficacy and resistance. The common physiological features of GBM tumors, such as hypoxia, hyaluronic acid, and glucose deprivation were found not to play a significant role in SMC efficacy. SMCs induced the death of microglia and macrophages, which are the major immune infiltrates in the tumor microenvironment. This death of microglia and macrophages then enhances the ability of SMCs to induce GBM cell death. Conversely, astrocytes promoted GBM cell growth and abrogated the ability of SMCs to induce death of GBM cells. The astrocyte-mediated resistance can be overcome in the presence of exogenous TNF-α. Overall, our results highlight that SMCs can induce death of microglia and macrophages, which then provides a source of death ligands for GBM cells, and that the targeting of astrocytes is a potential mechanism for overcoming SMC resistance for the treatment of GBM.
Collapse
Affiliation(s)
- Kyle Malone
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Melanie Dugas
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Nathalie Earl
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Tommy Alain
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Eric C LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Shawn T Beug
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
20
|
Chien F, Michaud ME, Bakhtiari M, Schroff C, Snuderl M, Velazquez Vega JE, MacDonald TJ, Bhasin MK. Medulloblastoma Spatial Transcriptomics Reveals Tumor Microenvironment Heterogeneity with High-Density Progenitor Cell Regions Correlating with High-Risk Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600684. [PMID: 38979174 PMCID: PMC11230370 DOI: 10.1101/2024.06.25.600684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The tumor microenvironment (TME) of medulloblastoma (MB) influences progression and therapy response, presenting a promising target for therapeutic advances. Prior single-cell analyses have characterized the cellular components of the TME but lack spatial context. To address this, we performed spatial transcriptomic sequencing on sixteen pediatric MB samples obtained at diagnosis, including two matched diagnosis-relapse pairs. Our analyses revealed inter- and intra-tumoral heterogeneity within the TME, comprised of tumor-associated astrocytes (TAAs), macrophages (TAMs), stromal components, and distinct subpopulations of MB cells at different stages of neuronal differentiation and cell cycle progression. We identified dense regions of quiescent progenitor-like MB cells enriched in patients with high-risk (HR) features and an increase in TAAs, TAMs, and dysregulated vascular endothelium following relapse. Our study presents novel insights into the spatial architecture and cellular landscape of the medulloblastoma TME, highlighting spatial patterns linked to HR features and relapse, which may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Franklin Chien
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, GA 30322, USA
| | - Marina E. Michaud
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Mojtaba Bakhtiari
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Chanel Schroff
- Department of Pathology, NYU Langone Health and Grossman School of Medicine, New York, NY 10016, USA
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health and Grossman School of Medicine, New York, NY 10016, USA
| | - Jose E. Velazquez Vega
- Department of Pathology and Laboratory Medicine, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, GA 30322, USA
| | - Tobey J. MacDonald
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, GA 30322, USA
| | - Manoj K. Bhasin
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
21
|
White J, White MPJ, Wickremesekera A, Peng L, Gray C. The tumour microenvironment, treatment resistance and recurrence in glioblastoma. J Transl Med 2024; 22:540. [PMID: 38844944 PMCID: PMC11155041 DOI: 10.1186/s12967-024-05301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024] Open
Abstract
The adaptability of glioblastoma (GBM) cells, encouraged by complex interactions with the tumour microenvironment (TME), currently renders GBM an incurable cancer. Despite intensive research, with many clinical trials, GBM patients rely on standard treatments including surgery followed by radiation and chemotherapy, which have been observed to induce a more aggressive phenotype in recurrent tumours. This failure to improve treatments is undoubtedly a result of insufficient models which fail to incorporate components of the human brain TME. Research has increasingly uncovered mechanisms of tumour-TME interactions that correlate to worsened patient prognoses, including tumour-associated astrocyte mitochondrial transfer, neuronal circuit remodelling and immunosuppression. This tumour hijacked TME is highly implicated in driving therapy resistance, with further alterations within the TME and tumour resulting from therapy exposure inducing increased tumour growth and invasion. Recent developments improving organoid models, including aspects of the TME, are paving an exciting future for the research and drug development for GBM, with the hopes of improving patient survival growing closer. This review focuses on GBMs interactions with the TME and their effect on tumour pathology and treatment efficiency, with a look at challenges GBM models face in sufficiently recapitulating this complex and highly adaptive cancer.
Collapse
Affiliation(s)
- Jasmine White
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | | | - Agadha Wickremesekera
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
22
|
Bugakova AS, Chudakova DA, Myzina MS, Yanysheva EP, Ozerskaya IV, Soboleva AV, Baklaushev VP, Yusubalieva GM. Non-Tumor Cells within the Tumor Microenvironment-The "Eminence Grise" of the Glioblastoma Pathogenesis and Potential Targets for Therapy. Cells 2024; 13:808. [PMID: 38786032 PMCID: PMC11119139 DOI: 10.3390/cells13100808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. GBM has high levels of therapy failure and its prognosis is usually dismal. The phenotypic heterogeneity of the tumor cells, dynamic complexity of non-tumor cell populations within the GBM tumor microenvironment (TME), and their bi-directional cross-talk contribute to the challenges of current therapeutic approaches. Herein, we discuss the etiology of GBM, and describe several major types of non-tumor cells within its TME, their impact on GBM pathogenesis, and molecular mechanisms of such an impact. We also discuss their value as potential therapeutic targets or prognostic biomarkers, with reference to the most recent works on this subject. We conclude that unless all "key player" populations of non-tumor cells within the TME are considered, no breakthrough in developing treatment for GBM can be achieved.
Collapse
Affiliation(s)
- Aleksandra S. Bugakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Maria S. Myzina
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Elvira P. Yanysheva
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Iuliia V. Ozerskaya
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Alesya V. Soboleva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
23
|
Li H, Zhu J, Liu X, Liu L, Huang S, Wu A, Xu Z, Zhang X, Li Z, Ni F, Liu L, Dong J. Glioma stem cell-derived exosomes induce the transformation of astrocytes via the miR-3065-5p/DLG2 signaling axis. Glia 2024; 72:857-871. [PMID: 38234042 DOI: 10.1002/glia.24506] [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: 02/19/2023] [Revised: 12/13/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Tumor-associated astrocytes (TAAs) in the glioblastoma microenvironment play an important role in tumor development and malignant progression initiated by glioma stem cells (GSCs). In the current study, normal human astrocytes (NHAs) were cultured and continuously treated with GSC-derived exosomes (GSC-EXOs) induction to explore the mechanism by which GSCs affect astrocyte remodeling. This study revealed that GSC-EXOs can induce the transformation of NHAs into TAAs, with relatively swollen cell bodies and multiple extended processes. In addition, high proliferation, elevated resistance to temozolomide (TMZ), and increased expression of TAA-related markers (TGF-β, CD44, and tenascin-C) were observed in the TAAs. Furthermore, GSC-derived exosomal miR-3065-5p could be delivered to NHAs, and miR-3065-5p levels increased significantly in TAAs, as verified by miRNA expression profile sequencing and Reverse transcription polymerase chain reaction. Overexpression of miR-3065-5p also enhanced NHA proliferation, elevated resistance to TMZ, and increased the expression levels of TAA-related markers. In addition, both GSC-EXO-induced and miR-3065-5p-overexpressing NHAs promoted tumorigenesis of GSCs in vivo. Discs Large Homolog 2 (DLG2, downregulated in glioblastoma) is a direct downstream target of miR-3065-5p in TAAs, and DLG2 overexpression could partially reverse the transformation of NHAs into TAAs. Collectively, these data demonstrate that GSC-EXOs induce the transformation of NHAs into TAAs via the miR-3065-5p/DLG2 signaling axis and that TAAs can further promote the tumorigenesis of GSCs. Thus, precisely blocking the interactions between astrocytes and GSCs via exosomes may be a novel strategy to inhibit glioblastoma development, but more in-depth mechanistic studies are still needed.
Collapse
Affiliation(s)
- Haoran Li
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianjun Zhu
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinglei Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Liu
- Department of Neurosurgery, The Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Shilu Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Anyi Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhipeng Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaopei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zengyang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fan Ni
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lijun Liu
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Wang M, Graner AN, Knowles B, McRae C, Fringuello A, Paucek P, Gavrilovic M, Redwine M, Hanson C, Coughlan C, Metzger B, Bolus V, Kopper T, Smith M, Zhou W, Lenz M, Abosch A, Ojemann S, Lillehei KO, Yu X, Graner MW. A tale of two tumors: differential, but detrimental, effects of glioblastoma extracellular vesicles (EVs) on normal human brain cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588622. [PMID: 38645117 PMCID: PMC11030303 DOI: 10.1101/2024.04.08.588622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Glioblastomas (GBMs) are dreadful brain tumors with abysmal survival outcomes. GBM EVs dramatically affect normal brain cells (largely astrocytes) constituting the tumor microenvironment (TME). EVs from different patient-derived GBM spheroids induced differential transcriptomic, secretomic, and proteomic effects on cultured astrocytes/brain tissue slices as GBM EV recipients. The net outcome of brain cell differential changes nonetheless converges on increased tumorigenicity. GBM spheroids and brain slices were derived from neurosurgical patient tissues following informed consent. Astrocytes were commercially obtained. EVs were isolated from conditioned culture media by ultrafiltration, ultraconcentration, and ultracentrifugation. EVs were characterized by nanoparticle tracking analysis, electron microscopy, biochemical markers, and proteomics. Astrocytes/brain tissues were treated with GBM EVs before downstream analyses. EVs from different GBMs induced brain cells to alter secretomes with pro-inflammatory or TME-modifying (proteolytic) effects. Astrocyte responses ranged from anti-viral gene/protein expression and cytokine release to altered extracellular signal-regulated protein kinase (ERK1/2) signaling pathways, and conditioned media from EV-treated cells increased GBM cell proliferation. Thus, astrocytes/brain slices treated with different GBM EVs underwent non-identical changes in various 'omics readouts and other assays, indicating "personalized" tumor-specific GBM EV effects on the TME. This raises concern regarding reliance on "model" systems as a sole basis for translational direction. Nonetheless, net downstream impacts from differential cellular and TME effects still led to increased tumorigenic capacities for the different GBMs.
Collapse
|
25
|
Yang H, Li M, Deng Y, Wen H, Luo M, Zhang W. Roles and interactions of tumor microenvironment components in medulloblastoma with implications for novel therapeutics. Genes Chromosomes Cancer 2024; 63:e23233. [PMID: 38607297 DOI: 10.1002/gcc.23233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 04/13/2024] Open
Abstract
Medulloblastomas, the most common malignant pediatric brain tumors, can be classified into the wingless, sonic hedgehog (SHH), group 3, and group 4 subgroups. Among them, the SHH subgroup with the TP53 mutation and group 3 generally present with the worst patient outcomes due to their high rates of recurrence and metastasis. A novel and effective treatment for refractory medulloblastomas is urgently needed. To date, the tumor microenvironment (TME) has been shown to influence tumor growth, recurrence, and metastasis through immunosuppression, angiogenesis, and chronic inflammation. Treatments targeting TME components have emerged as promising approaches to the treatment of solid tumors. In this review, we summarize progress in research on medulloblastoma microenvironment components and their interactions. We also discuss challenges and future research directions for TME-targeting medulloblastoma therapy.
Collapse
Affiliation(s)
- Hanjie Yang
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Min Li
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhao Deng
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huantao Wen
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Minjie Luo
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wangming Zhang
- Department of Pediatric Neurosurgery, Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Virtuoso A, D’Amico G, Scalia F, De Luca C, Papa M, Maugeri G, D’Agata V, Caruso Bavisotto C, D’Amico AG. The Interplay between Glioblastoma Cells and Tumor Microenvironment: New Perspectives for Early Diagnosis and Targeted Cancer Therapy. Brain Sci 2024; 14:331. [PMID: 38671983 PMCID: PMC11048111 DOI: 10.3390/brainsci14040331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) stands out as the most tremendous brain tumor, constituting 60% of primary brain cancers, accompanied by dismal survival rates. Despite advancements in research, therapeutic options remain limited to chemotherapy and surgery. GBM molecular heterogeneity, the intricate interaction with the tumor microenvironment (TME), and non-selective treatments contribute to the neoplastic relapse. Diagnostic challenges arise from GBM advanced-stage detection, necessitating the exploration of novel biomarkers for early diagnosis. Using data from the literature and a bioinformatic tool, the current manuscript delineates the molecular interplay between human GBM, astrocytes, and myeloid cells, underscoring selected protein pathways belonging to astroglia and myeloid lineage, which can be considered for targeted therapies. Moreover, the pivotal role of extracellular vesicles (EVs) in orchestrating a favorable microenvironment for cancer progression is highlighted, suggesting their utility in identifying biomarkers for GBM early diagnosis.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Giuseppa D’Amico
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Federica Scalia
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Ciro De Luca
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Velia D’Agata
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Agata Grazia D’Amico
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
| |
Collapse
|
27
|
Monteran L, Zait Y, Erez N. It's all about the base: stromal cells are central orchestrators of metastasis. Trends Cancer 2024; 10:208-229. [PMID: 38072691 DOI: 10.1016/j.trecan.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 03/16/2024]
Abstract
The tumor microenvironment (TME) is an integral part of tumors and plays a central role in all stages of carcinogenesis and progression. Each organ has a unique and heterogeneous microenvironment, which affects the ability of disseminated cells to grow in the new and sometimes hostile metastatic niche. Resident stromal cells, such as fibroblasts, osteoblasts, and astrocytes, are essential culprits in the modulation of metastatic progression: they transition from being sentinels of tissue integrity to being dysfunctional perpetrators that support metastatic outgrowth. Therefore, better understanding of the complexity of their reciprocal interactions with cancer cells and with other components of the TME is essential to enable the design of novel therapeutic approaches to prevent metastatic relapse.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Zait
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
28
|
Ballestín A, Armocida D, Ribecco V, Seano G. Peritumoral brain zone in glioblastoma: biological, clinical and mechanical features. Front Immunol 2024; 15:1347877. [PMID: 38487525 PMCID: PMC10937439 DOI: 10.3389/fimmu.2024.1347877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma is a highly aggressive and invasive tumor that affects the central nervous system (CNS). With a five-year survival rate of only 6.9% and a median survival time of eight months, it has the lowest survival rate among CNS tumors. Its treatment consists of surgical resection, subsequent fractionated radiotherapy and concomitant and adjuvant chemotherapy with temozolomide. Despite the implementation of clinical interventions, recurrence is a common occurrence, with over 80% of cases arising at the edge of the resection cavity a few months after treatment. The high recurrence rate and location of glioblastoma indicate the need for a better understanding of the peritumor brain zone (PBZ). In this review, we first describe the main radiological, cellular, molecular and biomechanical tissue features of PBZ; and subsequently, we discuss its current clinical management, potential local therapeutic approaches and future prospects.
Collapse
Affiliation(s)
- Alberto Ballestín
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Daniele Armocida
- Human Neurosciences Department, Neurosurgery Division, Sapienza University, Rome, Italy
| | - Valentino Ribecco
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Giorgio Seano
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| |
Collapse
|
29
|
Repici A, Ardizzone A, De Luca F, Colarossi L, Prestifilippo A, Pizzino G, Paterniti I, Esposito E, Capra AP. Signaling Pathways of AXL Receptor Tyrosine Kinase Contribute to the Pathogenetic Mechanisms of Glioblastoma. Cells 2024; 13:361. [PMID: 38391974 PMCID: PMC10886920 DOI: 10.3390/cells13040361] [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: 01/16/2024] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024] Open
Abstract
Brain tumors are a diverse collection of neoplasms affecting the brain with a high prevalence rate in people of all ages around the globe. In this pathological context, glioblastoma, a form of glioma that belongs to the IV-grade astrocytoma group, is the most common and most aggressive form of the primary brain tumors. Indeed, despite the best treatments available including surgery, radiotherapy or a pharmacological approach with Temozolomide, glioblastoma patients' mortality is still high, within a few months of diagnosis. Therefore, to increase the chances of these patients surviving, it is critical to keep finding novel treatment opportunities. In the past, efforts to treat glioblastoma have mostly concentrated on customized treatment plans that target specific mutations such as epidermal growth factor receptor (EGFR) mutations, Neurotrophic Tyrosine Receptor Kinase (NTRK) fusions, or multiple receptors using multi-kinase inhibitors like Sunitinib and Regorafenib, with varying degrees of success. Here, we focused on the receptor tyrosine kinase AXL that has been identified as a mediator for tumor progression and therapy resistance in various cancer types, including squamous cell tumors, small cell lung cancer, and breast cancer. Activated AXL leads to a significant increase in tumor proliferation, tumor cell migration, and angiogenesis in different in vitro and in vivo models of cancer since this receptor regulates interplay with apoptotic, angiogenic and inflammatory pathways. Based on these premises, in this review we mainly focused on the role of AXL in the course of glioblastoma, considering its primary biological mechanisms and as a possible target for the application of the most recent treatments.
Collapse
Affiliation(s)
- Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.R.); (A.A.); (F.D.L.); (I.P.); (A.P.C.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.R.); (A.A.); (F.D.L.); (I.P.); (A.P.C.)
| | - Fabiola De Luca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.R.); (A.A.); (F.D.L.); (I.P.); (A.P.C.)
| | - Lorenzo Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (L.C.); (A.P.); (G.P.)
| | - Angela Prestifilippo
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (L.C.); (A.P.); (G.P.)
| | - Gabriele Pizzino
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (L.C.); (A.P.); (G.P.)
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.R.); (A.A.); (F.D.L.); (I.P.); (A.P.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.R.); (A.A.); (F.D.L.); (I.P.); (A.P.C.)
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.R.); (A.A.); (F.D.L.); (I.P.); (A.P.C.)
| |
Collapse
|
30
|
Heinrich MA, Huynh NT, Heinrich L, Prakash J. Understanding glioblastoma stromal barriers against NK cell attack using tri-culture 3D spheroid model. Heliyon 2024; 10:e24808. [PMID: 38317968 PMCID: PMC10838749 DOI: 10.1016/j.heliyon.2024.e24808] [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: 07/18/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Glioblastoma multiforme (GBM), a highly aggressive tumor type with a dismal survival rate, has a poor outcome which is at least partly attributed to the crosstalk between cancer cells and cells from the tumor microenvironment such as astrocytes and microglia. We aimed to decipher the effect of these cells on GBM progression and on cell-based therapies using 3D co-cultures. Co-culturing of glioblastoma cells with patient-derived astrocytes or microglia or both formed dense and heterogeneous spheroids. Both, astrocytes and microglia, enhanced the spheroid growth rate and formed a physical barrier for macromolecules penetration, while only astrocytes enhanced the migration. Interestingly bi-/tri-cultured spheroids showed significant resistance against NK-92 cells, likely attributed to dense stroma and induced expression of immunosuppressive genes such as IDO1 or PTGES2. Altogether, our novel 3D GBM spheroid model recapitulates the cell-to-cell interactions of human glioblastoma and can serve as a suitable platform for evaluating cancer therapeutics.
Collapse
Affiliation(s)
| | | | - Lena Heinrich
- Department of Advanced Organ Bioengineering & Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500AE, Enschede, the Netherlands
| | - Jai Prakash
- Department of Advanced Organ Bioengineering & Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500AE, Enschede, the Netherlands
| |
Collapse
|
31
|
Dai J, Jiang Y, Hu H, Zhang S, Chen Y. Extracellular vesicles as modulators of glioblastoma progression and tumor microenvironment. Pathol Oncol Res 2024; 30:1611549. [PMID: 38379858 PMCID: PMC10876843 DOI: 10.3389/pore.2024.1611549] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Glioblastoma is the most aggressive brain tumor with extremely poor prognosis in adults. Routine treatments include surgery, chemotherapy, and radiotherapy; however, these may lead to rapid relapse and development of therapy-resistant tumor. Glioblastoma cells are known to communicate with macrophages, microglia, endothelial cells, astrocytes, and immune cells in the tumor microenvironment (TME) to promote tumor preservation. It was recently demonstrated that Glioblastoma-derived extracellular vesicles (EVs) participate in bidirectional intercellular communication in the TME. Apart from promoting glioblastoma cell proliferation, migration, and angiogenesis, EVs and their cargos (primarily proteins and miRNAs) can act as biomarkers for tumor diagnosis and prognosis. Furthermore, they can be used as therapeutic tools. In this review, the mechanisms of Glioblastoma-EVs biogenesis and intercellular communication with TME have been summarized. Moreover, there is discussion surrounding EVs as novel diagnostic structures and therapeutic tools for glioblastoma. Finally, unclear questions that require future investigation have been reviewed.
Collapse
Affiliation(s)
- Jie Dai
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Jiang
- Department of Neurosurgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyue Hu
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shuang Zhang
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yue Chen
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Liu X, Liu L, Wu A, Huang S, Xu Z, Zhang X, Li Z, Li H, Dong J. Transformed astrocytes confer temozolomide resistance on glioblastoma via delivering ALKBH7 to enhance APNG expression after educating by glioblastoma stem cells-derived exosomes. CNS Neurosci Ther 2024; 30:e14599. [PMID: 38332576 PMCID: PMC10853646 DOI: 10.1111/cns.14599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 11/01/2023] [Accepted: 12/20/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Glioblastoma is the most malignant primary brain tumor in adults. Temozolomide (TMZ) stands for the first-line chemotherapeutic agent against glioblastoma. Nevertheless, the therapeutic efficacy of TMZ appears to be remarkably limited, because of low cytotoxic efficiency against glioblastoma. Besides, various mechanical studies and the corresponding strategies fail to enhancing TMZ curative effect in clinical practice. Our previous studies have disclosed remodeling of glial cells by GSCs, but the roles of these transformed cells on promoting TMZ resistance have never been explored. METHODS Exosomes were extracted from GSCs culture through standard centrifugation procedures, which can activate transformation of normal human astrocytes (NHAs) totumor-associated astrocytes (TAAs) for 3 days through detect the level of TGF-β, CD44 and tenascin-C. The secretive protein level of ALKBH7 of TAAs was determined by ELISA kit. The protein level of APNG and ALKBH7 of GBM cells were determined by Western blot. Cell-based assays of ALKBH7 and APNG triggered drug resistance were performed through flow cytometric assay, Western blotting and colony formation assay respectively. A xenograft tumor model was applied to investigate the function of ALKBH7 in vivo. Finally, the effect of the ALKBH7/APNG signaling on TMZ resistance were evaluated by functional experiments. RESULTS Exosomes derived from GSCs can activate transformation of normal human astrocytes (NHAs)to tumor-associated astrocytes (TAAs), as well as up-regulation of ALKBH7expression in TAAs. Besides, TAAs derived ALKBH7 can regulate APNG gene expression of GBM cells. After co-culturing with TAAs for 5 days, ALKBH7 and APNG expression in GBM cells were elevated. Furthermore, Knocking-down of APNG increased the inhibitory effect of TMZ on GBM cells survival. CONCLUSION The present study illustrated a new mechanism of glioblastoma resistance to TMZ, which based on GSCs-exo educated TAAs delivering ALKBH7 to enhance APNG expression of GBM cells, which implied that targeting on ALKBH7/APNG regulation network may provide a new strategy of enhancing TMZ therapeutic effects against glioblastoma.
Collapse
Affiliation(s)
- Xinglei Liu
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Anyi Wu
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Shilu Huang
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhipeng Xu
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiaopei Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zengyang Li
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Haoran Li
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jun Dong
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
33
|
Boyineni J, Wood JM, Ravindra A, Boley E, Donohue SE, Soares MB, Malchenko S. Prospective Approach to Deciphering the Impact of Intercellular Mitochondrial Transfer from Human Neural Stem Cells and Brain Tumor-Initiating Cells to Neighboring Astrocytes. Cells 2024; 13:204. [PMID: 38334595 PMCID: PMC10854889 DOI: 10.3390/cells13030204] [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: 12/05/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/10/2024] Open
Abstract
The communication between neural stem cells (NSCs) and surrounding astrocytes is essential for the homeostasis of the NSC niche. Intercellular mitochondrial transfer, a unique communication system that utilizes the formation of tunneling nanotubes for targeted mitochondrial transfer between donor and recipient cells, has recently been identified in a wide range of cell types. Intercellular mitochondrial transfer has also been observed between different types of cancer stem cells (CSCs) and their neighboring cells, including brain CSCs and astrocytes. CSC mitochondrial transfer significantly enhances overall tumor progression by reprogramming neighboring cells. Despite the urgent need to investigate this newly identified phenomenon, mitochondrial transfer in the central nervous system remains largely uncharacterized. In this study, we found evidence of intercellular mitochondrial transfer from human NSCs and from brain CSCs, also known as brain tumor-initiating cells (BTICs), to astrocytes in co-culture experiments. Both NSC and BTIC mitochondria triggered similar transcriptome changes upon transplantation into the recipient astrocytes. In contrast to NSCs, the transplanted mitochondria from BTICs had a significant proliferative effect on the recipient astrocytes. This study forms the basis for mechanistically deciphering the impact of intercellular mitochondrial transfer on recipient astrocytes, which will potentially provide us with new insights into the mechanisms of mitochondrial retrograde signaling.
Collapse
Affiliation(s)
- Jerusha Boyineni
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| | - Jason Michael Wood
- Research Informatics Core, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Aditya Ravindra
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| | - Ethan Boley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| | - Sarah E. Donohue
- Research Services, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA;
| | - Marcelo Bento Soares
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
- Department of Psychiatry & Behavioral Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Sergey Malchenko
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| |
Collapse
|
34
|
Yadav N, Purow BW. Understanding current experimental models of glioblastoma-brain microenvironment interactions. J Neurooncol 2024; 166:213-229. [PMID: 38180686 PMCID: PMC11056965 DOI: 10.1007/s11060-023-04536-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024]
Abstract
Glioblastoma (GBM) is a common and devastating primary brain tumor, with median survival of 16-18 months after diagnosis in the setting of substantial resistance to standard-of-care and inevitable tumor recurrence. Recent work has implicated the brain microenvironment as being critical for GBM proliferation, invasion, and resistance to treatment. GBM does not operate in isolation, with neurons, astrocytes, and multiple immune populations being implicated in GBM tumor progression and invasiveness. The goal of this review article is to provide an overview of the available in vitro, ex vivo, and in vivo experimental models for assessing GBM-brain interactions, as well as discuss each model's relative strengths and limitations. Current in vitro models discussed will include 2D and 3D co-culture platforms with various cells of the brain microenvironment, as well as spheroids, whole organoids, and models of fluid dynamics, such as interstitial flow. An overview of in vitro and ex vivo organotypic GBM brain slices is also provided. Finally, we conclude with a discussion of the various in vivo rodent models of GBM, including xenografts, syngeneic grafts, and genetically-engineered models of GBM.
Collapse
Affiliation(s)
- Niket Yadav
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Benjamin W Purow
- Department of Neurology, University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA, 22903, USA.
| |
Collapse
|
35
|
Popov A, Brazhe N, Morozova K, Yashin K, Bychkov M, Nosova O, Sutyagina O, Brazhe A, Parshina E, Li L, Medyanik I, Korzhevskii DE, Shenkarev Z, Lyukmanova E, Verkhratsky A, Semyanov A. Mitochondrial malfunction and atrophy of astrocytes in the aged human cerebral cortex. Nat Commun 2023; 14:8380. [PMID: 38104196 PMCID: PMC10725430 DOI: 10.1038/s41467-023-44192-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
How aging affects cells of the human brain active milieu remains largely unknown. Here, we analyze astrocytes and neurons in the neocortical tissue of younger (22-50 years) and older (51-72 years) adults. Aging decreases the amount of reduced mitochondrial cytochromes in astrocytes but not neurons. The protein-to-lipid ratio decreases in astrocytes and increases in neurons. Aged astrocytes show morphological atrophy quantified by the decreased length of branches, decreased volume fraction of leaflets, and shrinkage of the anatomical domain. Atrophy correlates with the loss of gap junction coupling between astrocytes and increased input resistance. Aging is accompanied by the upregulation of glial fibrillary acidic protein (GFAP) and downregulation of membrane-cytoskeleton linker ezrin associated with leaflets. No significant changes in neuronal excitability or spontaneous inhibitory postsynaptic signaling is observed. Thus, brain aging is associated with the impaired morphological presence and mitochondrial malfunction of cortical astrocytes, but not neurons.
Collapse
Affiliation(s)
- Alexander Popov
- College of Medicine, Jiaxing University, 314001, Jiaxing, Zhejiang Pro, China
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Nadezda Brazhe
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Kseniia Morozova
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Konstantin Yashin
- Department of Neurosurgery, Privolzhskiy Research Medical University, Nizhny, Novgorod, 603005, Russia
| | - Maxim Bychkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Olga Nosova
- Institute of Experimental Medicine, St. Petersburg, 197376, Russia
| | - Oksana Sutyagina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Alexey Brazhe
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Evgenia Parshina
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Li Li
- College of Medicine, Jiaxing University, 314001, Jiaxing, Zhejiang Pro, China
| | - Igor Medyanik
- Department of Neurosurgery, Privolzhskiy Research Medical University, Nizhny, Novgorod, 603005, Russia
| | | | - Zakhar Shenkarev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Ekaterina Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
- Faculty of Biology, Shenzhen MSU-BIT University, 518172, Shenzhen, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain.
| | - Alexey Semyanov
- College of Medicine, Jiaxing University, 314001, Jiaxing, Zhejiang Pro, China.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia.
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia.
- Sechenov First Moscow State Medical University, Moscow, 119435, Russia.
| |
Collapse
|
36
|
Lia A, Di Spiezio A, Vitalini L, Tore M, Puja G, Losi G. Ion Channels and Ionotropic Receptors in Astrocytes: Physiological Functions and Alterations in Alzheimer's Disease and Glioblastoma. Life (Basel) 2023; 13:2038. [PMID: 37895420 PMCID: PMC10608464 DOI: 10.3390/life13102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
The human brain is composed of nearly one hundred billion neurons and an equal number of glial cells, including macroglia, i.e., astrocytes and oligodendrocytes, and microglia, the resident immune cells of the brain. In the last few decades, compelling evidence has revealed that glial cells are far more active and complex than previously thought. In particular, astrocytes, the most abundant glial cell population, not only take part in brain development, metabolism, and defense against pathogens and insults, but they also affect sensory, motor, and cognitive functions by constantly modulating synaptic activity. Not surprisingly, astrocytes are actively involved in neurodegenerative diseases (NDs) and other neurological disorders like brain tumors, in which they rapidly become reactive and mediate neuroinflammation. Reactive astrocytes acquire or lose specific functions that differently modulate disease progression and symptoms, including cognitive impairments. Astrocytes express several types of ion channels, including K+, Na+, and Ca2+ channels, transient receptor potential channels (TRP), aquaporins, mechanoreceptors, and anion channels, whose properties and functions are only partially understood, particularly in small processes that contact synapses. In addition, astrocytes express ionotropic receptors for several neurotransmitters. Here, we provide an extensive and up-to-date review of the roles of ion channels and ionotropic receptors in astrocyte physiology and pathology. As examples of two different brain pathologies, we focus on Alzheimer's disease (AD), one of the most diffuse neurodegenerative disorders, and glioblastoma (GBM), the most common brain tumor. Understanding how ion channels and ionotropic receptors in astrocytes participate in NDs and tumors is necessary for developing new therapeutic tools for these increasingly common neurological conditions.
Collapse
Affiliation(s)
- Annamaria Lia
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
| | - Alessandro Di Spiezio
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
- Neuroscience Institute (CNR-IN), Padova Section, 35131 Padova, Italy
| | - Lorenzo Vitalini
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Manuela Tore
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Puja
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Gabriele Losi
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
37
|
Gonçalves TL, de Araújo LP, Pereira Ferrer V. Tamoxifen as a modulator of CXCL12-CXCR4-CXCR7 chemokine axis: A breast cancer and glioblastoma view. Cytokine 2023; 170:156344. [PMID: 37639844 DOI: 10.1016/j.cyto.2023.156344] [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: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
The chemokine stromal cell-derived-factor 1 (SDF)-1/CXCL12 acts by binding to its receptors, the CXC-4 chemokine receptor (CXCR4) and the CXC-7 chemokine receptor (CXCR7). The binding of CXCL12 to its receptors results in downstream signaling that leads to cell survival, proliferation and migration of tumor cells. CXCL12 and CXCR4 are highly expressed in breast cancer (BC) and glioblastoma (GBM) compared to normal cells. High expression of this chemokine axis correlates with increased therapy resistance and grade, tumor spread and poorer prognosis in these tumors. Tamoxifen (TMX) is a selective estrogen receptor modulator (SERM) that inhibits the expression of estrogen-regulated genes, including growth and angiogenic factors secreted by tumor cells. Additionally, TMX targets several proteins, such as protein kinase C (PKC), phospholipase C (PLC), P-glycoprotein (PgP), phosphatidylinositol-3-kinase (PI3K) and ion channels. This drug showed promising antitumor activity against both BC and GBM cells. In this review, we discuss the role of the CXCL12-CXCR4-CXCR7 chemokine axis in BC and GBM tumor biology and propose TMX as a potential modulator of this axis in these tumors. TMX modulates the CXCL12-CXCR4-CXCR7 axis in BC, however, there are no studies on this in GBM. We propose that studying this axis in GBM cells/patients treated with TMX might be beneficial for these patients. TMX inhibits important signaling pathways in these tumors and the activation of this chemokine axis is associated with increased therapy resistance.
Collapse
Affiliation(s)
- Thaynan Lopes Gonçalves
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Postgraduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Luanna Prudencio de Araújo
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Valéria Pereira Ferrer
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Postgraduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil.
| |
Collapse
|
38
|
Duan W, Xia S, Tang M, Lin M, Liu W, Wang Q. Targeting of endothelial cells in brain tumours. Clin Transl Med 2023; 13:e1433. [PMID: 37830128 PMCID: PMC10570772 DOI: 10.1002/ctm2.1433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/11/2023] [Accepted: 09/30/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Aggressive brain tumours, whether primary gliomas or secondary metastases, are characterised by hypervascularisation and are fatal. Recent research has emphasised the crucial involvement of endothelial cells (ECs) in all brain tumour genesis and development events, with various patterns and underlying mechanisms identified. MAIN BODY Here, we highlight recent advances in knowledge about the contributions of ECs to brain tumour development, providing a comprehensive summary including descriptions of interactions between ECs and tumour cells, the heterogeneity of ECs and new models for research on ECs in brain malignancies. We also discuss prospects for EC targeting in novel therapeutic approaches. CONCLUSION Interventions targeting ECs, as an adjunct to other therapies (e.g. immunotherapies, molecular-targeted therapies), have shown promising clinical efficacy due to the high degree of vascularisation in brain tumours. Developing precise strategies to target tumour-associated vessels based on the heterogeneity of ECs is expected to improve anti-vascular efficacy.
Collapse
Affiliation(s)
- Wenzhe Duan
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Shengkai Xia
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Mengyi Tang
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Manqing Lin
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Wenwen Liu
- Cancer Translational Medicine Research CenterThe Second HospitalDalian Medical UniversityDalianChina
| | - Qi Wang
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
- Cancer Translational Medicine Research CenterThe Second HospitalDalian Medical UniversityDalianChina
| |
Collapse
|
39
|
Cui Y, Lee P, Reardon JJ, Wang A, Lynch S, Otero JJ, Sizemore G, Winter JO. Evaluating glioblastoma tumour sphere growth and migration in interaction with astrocytes using 3D collagen-hyaluronic acid hydrogels. J Mater Chem B 2023; 11:5442-5459. [PMID: 37159233 PMCID: PMC10330682 DOI: 10.1039/d3tb00066d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Glioblastoma (GB) is an astrocytic brain tumour with a low survival rate, partly because of its highly invasive nature. The GB tumour microenvironment (TME) includes its extracellular matrix (ECM), a variety of brain cell types, unique anatomical structures, and local mechanical cues. As such, researchers have attempted to create biomaterials and culture models that mimic features of TME complexity. Hydrogel materials have been particularly popular because they enable 3D cell culture and mimic TME mechanical properites and chemical composition. Here, we used a 3D collagen I-hyaluronic acid hydrogel material to explore interactions between GB cells and astrocytes, the normal cell type from which GB likely derives. We demonstrate three different spheroid culture configurations, including GB multi-spheres (i.e., GB and astrocyte cells in spheroid co-culture), GB-only mono-spheres cultured with astrocyte-conditioned media, and GB-only mono-spheres cultured with dispersed live or fixed astrocytes. Using U87 and LN229 GB cell lines and primary human astrocytes, we investigated material and experiment variability. We then used time-lapse fluorescence microscopy to measure invasive potential by characterizing the sphere size, migration capacity, and weight-averaged migration distance in these hydrogels. Finally, we developed methods to extract RNA for gene expression analysis from cells cultured in hydrogels. U87 and LN229 cells displayed different migration behaviors. U87 migration occurred primarily as single cells and was reduced with higher numbers of astrocytes in both multi-sphere and mono-sphere plus dispersed astrocyte cultures. In contrast, LN229 migration exhibited features of collective migration and was increased in monosphere plus dispersed astrocyte cultures. Gene expression studies indicated that the most differentially expressed genes in these co-cultures were CA9, HLA-DQA1, TMPRSS2, FPR1, OAS2, and KLRD1. Most differentially expressed genes were related to immune response, inflammation, and cytokine signalling, with greater influence on U87 than LN229. These data show that 3D in vitro hydrogel co-culture models can be used to reveal cell line specific differences in migration and to study differential GB-astrocyte crosstalk.
Collapse
Affiliation(s)
- Yixiao Cui
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| | - Paul Lee
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Jesse J Reardon
- Ohio State University Comprehensive Cancer Center - James, The Ohio State University, Columbus, OH, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, OH, USA
| | - Anna Wang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| | - Skylar Lynch
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| | - Jose J Otero
- Ohio State University Comprehensive Cancer Center - James, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Gina Sizemore
- Ohio State University Comprehensive Cancer Center - James, The Ohio State University, Columbus, OH, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, OH, USA
| | - Jessica O Winter
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
- Ohio State University Comprehensive Cancer Center - James, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
40
|
Cheng Y, Li S, Hou Y, Wang W, Wang K, Fu S, Yuan Y, Yang K, Ye X. Glioma-derived small extracellular vesicles induce pericyte-phenotype transition of glioma stem cells under hypoxic conditions. Cell Signal 2023:110754. [PMID: 37315748 DOI: 10.1016/j.cellsig.2023.110754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and lethal primary brain tumor characterized by extensive vascularization. Anti-angiogenic therapy for this cancer offers the possibility of universal efficacy. However, preclinical and clinical studies suggest that anti-VEGF drug such as Bevacizumab actively promotes tumor invasion, which ultimately leads to a therapy-resistant and recurrent phenotype of GBMs. Whether Bevacizumab can improve survival over chemotherapy alone remains debated. Herein, we emphasized the importance of small extracellular vesicles (sEVs) internalization by glioma stem cells (GSCs) in giving rise to the failure of anti-angiogenic therapy in the treatment of GBMs and discovered a specific therapeutic target for this damaging disease. METHODS To experimentally prove that hypoxia condition promotes the release of GBM cells-derived sEVs, which could be taken up by the surrounding GSCs, we used an ultracentrifugation strategy to isolate GBM-derived sEVs under hypoxic or normoxic conditions, performed bioinformatics analysis and multidimensional molecular biology experiments, and established a xenograft mouse model. RESULTS The internalization of sEVs by GSCs was proved to promote tumor growth and angiogenesis through the pericyte-phenotype transition. Hypoxia-derived sEVs could efficiently deliver TGF-β1 to GSCs, thus resulting in the activation of the TGF-β signaling pathway and the consequent pericyte-phenotype transition. Specifically targeting GSC-derived pericyte using Ibrutinib can reverse the effects of GBM-derived sEVs and enhance the tumor-eradicating effects when combined with Bevacizumab. CONCLUSION This present study provides a new interpretation of the failure of anti-angiogenic therapy in the non-operative treatment of GBMs and discovers a promising therapeutic target for this intractable disease.
Collapse
Affiliation(s)
- Yue Cheng
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Shijie Li
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Yongying Hou
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Weijun Wang
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Ke Wang
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Shihui Fu
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan Province, PR China
| | - Ye Yuan
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, PR China.
| | - Kaidi Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; Department of Oncology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan Province, PR China.
| | - Xiufeng Ye
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China.
| |
Collapse
|
41
|
Zheng C, Mao C, Tang K, Shu H. VSIG4 Silencing Inhibits Glioblastoma Growth by Regulating the JAK2/STAT3 Pathway. Neuropsychiatr Dis Treat 2023; 19:1397-1408. [PMID: 37292180 PMCID: PMC10246575 DOI: 10.2147/ndt.s406782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023] Open
Abstract
Background Glioblastoma (GBM) is the most common malignant primary brain tumour in adults. VSIG4 has been identified to be associated with GBM. We aimed to determine the downstream regulatory mechanisms of VSIG4 in GBM. Methods Differential expression of VSIG4 was analysed using GEPIA. The expression of VSIG4 was assessed by RT-qPCR and its downstream genes were screened by transcriptome sequencing. The expression of pyroptosis-related proteins and the JAK2/STAT3 pathway was measured by Western blotting. GBM cell viability, migration, and invasion were detected using CCK-8, scratch, and Transwell assays. The levels of pyroptosis-related factors were measured using ELISA. The effect of VSIG4 on GBM tumour growth in vivo was explored by constructing a xenograft tumour model. Results VSIG4 expression was upregulated in GBM. Functionally, silencing of VSIG4 inhibited proliferation, invasion, and migration of U251 and LN229 cells, and promoted pyroptosis. Mechanically, transcriptome sequencing revealed that the JAK2/STAT3 pathway might be a downstream regulator of VSIG4. Further studies proved that silencing of VSIG4 enhanced the expression of p-JAK2 and p-STAT3, and the JAK2/STAT3 pathway inhibitor relieved the suppression of VSIG4 silencing on GBM cell viability, invasion, and migration. Furthermore, in vivo experiments further validated that knockdown of VSIG4 inhibited the growth of GBM tumors. Conclusion In GBM, silencing VSIG4 promoted pyroptosis and inhibited tumor progression by regulating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Congying Zheng
- Department of Neurosurgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Chengliang Mao
- Department of Neurosurgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Kai Tang
- Department of Neurosurgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Hang Shu
- Department of Neurosurgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| |
Collapse
|
42
|
Lim J, Kang I, La J, Ku KB, Kang BH, Kim Y, Park WH, Lee HK. Harnessing type I interferon-mediated immunity to target malignant brain tumors. Front Immunol 2023; 14:1203929. [PMID: 37304294 PMCID: PMC10247981 DOI: 10.3389/fimmu.2023.1203929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Type I interferons have long been appreciated as a cytokine family that regulates antiviral immunity. Recently, their role in eliciting antitumor immune responses has gained increasing attention. Within the immunosuppressive tumor microenvironment (TME), interferons stimulate tumor-infiltrating lymphocytes to promote immune clearance and essentially reshape a "cold" TME into an immune-activating "hot" TME. In this review, we focus on gliomas, with an emphasis on malignant glioblastoma, as these brain tumors possess a highly invasive and heterogenous brain TME. We address how type I interferons regulate antitumor immune responses against malignant gliomas and reshape the overall immune landscape of the brain TME. Furthermore, we discuss how these findings can translate into future immunotherapies targeting brain tumors in general.
Collapse
Affiliation(s)
- Juhee Lim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - In Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jeongwoo La
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Keun Bon Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Byeong Hoon Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yumin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Won Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
43
|
Yuile A, Wei JQ, Mohan AA, Hotchkiss KM, Khasraw M. Interdependencies of the Neuronal, Immune and Tumor Microenvironment in Gliomas. Cancers (Basel) 2023; 15:2856. [PMID: 37345193 PMCID: PMC10216320 DOI: 10.3390/cancers15102856] [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: 02/10/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Gliomas are the most common primary brain malignancy and are universally fatal. Despite significant breakthrough in understanding tumor biology, treatment breakthroughs have been limited. There is a growing appreciation that major limitations on effective treatment are related to the unique and highly complex glioma tumor microenvironment (TME). The TME consists of multiple different cell types, broadly categorized into tumoral, immune and non-tumoral, non-immune cells. Each group provides significant influence on the others, generating a pro-tumor dynamic with significant immunosuppression. In addition, glioma cells are highly heterogenous with various molecular distinctions on the cellular level. These variations, in turn, lead to their own unique influence on the TME. To develop future treatments, an understanding of this complex TME interplay is needed. To this end, we describe the TME in adult gliomas through interactions between its various components and through various glioma molecular phenotypes.
Collapse
Affiliation(s)
- Alexander Yuile
- Department of Medical Oncology, Royal North Shore Hospital, Reserve Road, St Leonards, NSW 2065, Australia
- The Brain Cancer Group, North Shore Private Hospital, 3 Westbourne Street, St Leonards, NSW 2065, Australia
- Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Joe Q. Wei
- Department of Medical Oncology, Royal North Shore Hospital, Reserve Road, St Leonards, NSW 2065, Australia
- Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Aditya A. Mohan
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| | - Kelly M. Hotchkiss
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| | - Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| |
Collapse
|
44
|
Hey G, Rao R, Carter A, Reddy A, Valle D, Patel A, Patel D, Lucke-Wold B, Pomeranz Krummel D, Sengupta S. Ligand-Gated Ion Channels: Prognostic and Therapeutic Implications for Gliomas. J Pers Med 2023; 13:853. [PMID: 37241023 PMCID: PMC10224160 DOI: 10.3390/jpm13050853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Gliomas are common primary brain malignancies that remain difficult to treat due to their overall aggressiveness and heterogeneity. Although a variety of therapeutic strategies have been employed for the treatment of gliomas, there is increasing evidence that suggests ligand-gated ion channels (LGICs) can serve as a valuable biomarker and diagnostic tool in the pathogenesis of gliomas. Various LGICs, including P2X, SYT16, and PANX2, have the potential to become altered in the pathogenesis of glioma, which can disrupt the homeostatic activity of neurons, microglia, and astrocytes, further exacerbating the symptoms and progression of glioma. Consequently, LGICs, including purinoceptors, glutamate-gated receptors, and Cys-loop receptors, have been targeted in clinical trials for their potential therapeutic benefit in the diagnosis and treatment of gliomas. In this review, we discuss the role of LGICs in the pathogenesis of glioma, including genetic factors and the effect of altered LGIC activity on the biological functioning of neuronal cells. Additionally, we discuss current and emerging investigations regarding the use of LGICs as a clinical target and potential therapeutic for gliomas.
Collapse
Affiliation(s)
- Grace Hey
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rohan Rao
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ashley Carter
- Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Akshay Reddy
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Daisy Valle
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Anjali Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Drashti Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 23608, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Soma Sengupta
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
45
|
Cao Y. Neural induction drives body axis formation during embryogenesis, but a neural induction-like process drives tumorigenesis in postnatal animals. Front Cell Dev Biol 2023; 11:1092667. [PMID: 37228646 PMCID: PMC10203556 DOI: 10.3389/fcell.2023.1092667] [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: 11/08/2022] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Characterization of cancer cells and neural stem cells indicates that tumorigenicity and pluripotency are coupled cell properties determined by neural stemness, and tumorigenesis represents a process of progressive loss of original cell identity and gain of neural stemness. This reminds of a most fundamental process required for the development of the nervous system and body axis during embryogenesis, i.e., embryonic neural induction. Neural induction is that, in response to extracellular signals that are secreted by the Spemann-Mangold organizer in amphibians or the node in mammals and inhibit epidermal fate in ectoderm, the ectodermal cells lose their epidermal fate and assume the neural default fate and consequently, turn into neuroectodermal cells. They further differentiate into the nervous system and also some non-neural cells via interaction with adjacent tissues. Failure in neural induction leads to failure of embryogenesis, and ectopic neural induction due to ectopic organizer or node activity or activation of embryonic neural genes causes a formation of secondary body axis or a conjoined twin. During tumorigenesis, cells progressively lose their original cell identity and gain of neural stemness, and consequently, gain of tumorigenicity and pluripotency, due to various intra-/extracellular insults in cells of a postnatal animal. Tumorigenic cells can be induced to differentiation into normal cells and integrate into normal embryonic development within an embryo. However, they form tumors and cannot integrate into animal tissues/organs in a postnatal animal because of lack of embryonic inducing signals. Combination of studies of developmental and cancer biology indicates that neural induction drives embryogenesis in gastrulating embryos but a similar process drives tumorigenesis in a postnatal animal. Tumorigenicity is by nature the manifestation of aberrant occurrence of pluripotent state in a postnatal animal. Pluripotency and tumorigenicity are both but different manifestations of neural stemness in pre- and postnatal stages of animal life, respectively. Based on these findings, I discuss about some confusion in cancer research, propose to distinguish the causality and associations and discriminate causal and supporting factors involved in tumorigenesis, and suggest revisiting the focus of cancer research.
Collapse
Affiliation(s)
- Ying Cao
- Shenzhen Research Institute of Nanjing University, Shenzhen, China
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
46
|
Chin SM, Reina G, Chau NDQ, Chabrol T, Wion D, Bouamrani A, Gay E, Nishina Y, Bianco A, Berger F. Functional Graphene for Peritumoral Brain Microenvironment Modulation Therapy in Glioblastoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208227. [PMID: 36732906 DOI: 10.1002/smll.202208227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/09/2023] [Indexed: 05/04/2023]
Abstract
Peritumoral brain invasion is the main target to cure glioblastoma. Chemoradiotherapy and targeted therapies fail to combat peritumoral relapse. Brain inaccessibility and tumor heterogeneity explain this failure, combined with overlooking the peritumor microenvironment. Reduce graphene oxide (rGO) provides a unique opportunity to modulate the local brain microenvironment. Multimodal graphene impacts are reported on glioblastoma cells in vitro but fail when translated in vivo because of low diffusion. This issue is solved by developing a new rGO formulation involving ultramixing during the functionalization with polyethyleneimine (PEI) leading to the formation of highly water-stable rGO-PEI. Wide mice brain diffusion and biocompatibility are demonstrated. Using an invasive GL261 model, an anti-invasive effect is observed. A major unexpected modification of the peritumoral area is also observed with the neutralization of gliosis. In vitro, mechanistic investigations are performed using primary astrocytes and cytokine array. The result suggests that direct contact of rGO-PEIUT neutralizes astrogliosis, decreasing several proinflammatory cytokines that would explain a bystander tumor anti-invasive effect. rGO also significantly downregulates several proinvasive/protumoral cytokines at the tumor cell level. The results open the way to a new microenvironment anti-invasive nanotherapy using a new graphene nanomaterial that is optimized for in vivo brain delivery.
Collapse
Affiliation(s)
- Shan Min Chin
- Emmanuel Gay, François Berger, INSERM UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Giacomo Reina
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Ngoc Do Quyen Chau
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Tanguy Chabrol
- Emmanuel Gay, François Berger, INSERM UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Didier Wion
- Emmanuel Gay, François Berger, INSERM UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Ali Bouamrani
- Emmanuel Gay, François Berger, INSERM UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Emmanuel Gay
- Emmanuel Gay, François Berger, INSERM UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| | - Yuta Nishina
- Graduate School of Natural Science and Technology, Okayama University, Tsushimanaka, Kita-ku, Okayama, 700-8530, Japan
- Research Core for Interdisciplinary Sciences, Okayama University, Tsushimanaka, Kita-ku, Okayama, 700-8530, Japan
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - François Berger
- Emmanuel Gay, François Berger, INSERM UMR1205, Brain Tech Lab, Grenoble Alpes University, Grenoble, 38000, France
| |
Collapse
|
47
|
Erices JI, Bizama C, Niechi I, Uribe D, Rosales A, Fabres K, Navarro-Martínez G, Torres Á, San Martín R, Roa JC, Quezada-Monrás C. Glioblastoma Microenvironment and Invasiveness: New Insights and Therapeutic Targets. Int J Mol Sci 2023; 24:7047. [PMID: 37108208 PMCID: PMC10139189 DOI: 10.3390/ijms24087047] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 04/29/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain cancer in adults. Without treatment the mean patient survival is approximately 6 months, which can be extended to 15 months with the use of multimodal therapies. The low effectiveness of GBM therapies is mainly due to the tumor infiltration into the healthy brain tissue, which depends on GBM cells' interaction with the tumor microenvironment (TME). The interaction of GBM cells with the TME involves cellular components such as stem-like cells, glia, endothelial cells, and non-cellular components such as the extracellular matrix, enhanced hypoxia, and soluble factors such as adenosine, which promote GBM's invasiveness. However, here we highlight the role of 3D patient-derived glioblastoma organoids cultures as a new platform for study of the modeling of TME and invasiveness. In this review, the mechanisms involved in GBM-microenvironment interaction are described and discussed, proposing potential prognosis biomarkers and new therapeutic targets.
Collapse
Affiliation(s)
- José Ignacio Erices
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Carolina Bizama
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Ignacio Niechi
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Daniel Uribe
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Arnaldo Rosales
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Karen Fabres
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Giovanna Navarro-Martínez
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Ángelo Torres
- Escuela de Medicina Veterinaria, Facultad de Recursos Naturales y Medicina Veterinaria, Universidad Santo Tomás, Talca 8370003, Chile
| | - Rody San Martín
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Claudia Quezada-Monrás
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| |
Collapse
|
48
|
Wu JR, Hernandez Y, Miyasaki KF, Kwon EJ. Engineered nanomaterials that exploit blood-brain barrier dysfunction fordelivery to the brain. Adv Drug Deliv Rev 2023; 197:114820. [PMID: 37054953 DOI: 10.1016/j.addr.2023.114820] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
The blood-brain barrier (BBB) is a highly regulated physical and functional boundarythat tightly controls the transport of materials between the blood and the brain. There is an increasing recognition that the BBB is dysfunctional in a wide range of neurological disorders; this dysfunction can be symptomatic of the disease but can also play a role in disease etiology. BBB dysfunction can be exploited for the delivery of therapeutic nanomaterials. Forexample, there can be a transient, physical disruption of the BBB in diseases such as brain injury and stroke, which allows temporary access of nanomaterials into the brain. Physicaldisruption of the BBB through external energy sources is now being clinically pursued toincrease therapeutic delivery into the brain. In other diseases, the BBB takes on new properties that can beleveraged by delivery carriers. For instance, neuroinflammation induces the expression ofreceptors on the BBB that can be targeted by ligand-modified nanomaterials and theendogenous homing of immune cells into the diseased brain can be hijacked for the delivery ofnanomaterials. Lastly, BBB transport pathways can be altered to increase nanomaterial transport. In this review, we will describe changes that can occur in the BBB in disease, and how these changes have been exploited by engineered nanomaterials forincreased transport into the brain.
Collapse
Affiliation(s)
- Jason R Wu
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Yazmin Hernandez
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Katelyn F Miyasaki
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Ester J Kwon
- Department of Bioengineering, University of California San Diego, La Jolla, CA; Sanford Consortium for Regenerative Medicine.
| |
Collapse
|
49
|
Caverzán MD, Beaugé L, Oliveda PM, Cesca González B, Bühler EM, Ibarra LE. Exploring Monocytes-Macrophages in Immune Microenvironment of Glioblastoma for the Design of Novel Therapeutic Strategies. Brain Sci 2023; 13:brainsci13040542. [PMID: 37190507 DOI: 10.3390/brainsci13040542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Gliomas are primary malignant brain tumors. These tumors seem to be more and more frequent, not only because of a true increase in their incidence, but also due to the increase in life expectancy of the general population. Among gliomas, malignant gliomas and more specifically glioblastomas (GBM) are a challenge in their diagnosis and treatment. There are few effective therapies for these tumors, and patients with GBM fare poorly, even after aggressive surgery, chemotherapy, and radiation. Over the last decade, it is now appreciated that these tumors are composed of numerous distinct tumoral and non-tumoral cell populations, which could each influence the overall tumor biology and response to therapies. Monocytes have been proved to actively participate in tumor growth, giving rise to the support of tumor-associated macrophages (TAMs). In GBM, TAMs represent up to one half of the tumor mass cells, including both infiltrating macrophages and resident brain microglia. Infiltrating macrophages/monocytes constituted ~ 85% of the total TAM population, they have immune functions, and they can release a wide array of growth factors and cytokines in response to those factors produced by tumor and non-tumor cells from the tumor microenvironment (TME). A brief review of the literature shows that this cell population has been increasingly studied in GBM TME to understand its role in tumor progression and therapeutic resistance. Through the knowledge of its biology and protumoral function, the development of therapeutic strategies that employ their recruitment as well as the modulation of their immunological phenotype, and even the eradication of the cell population, can be harnessed for therapeutic benefit. This revision aims to summarize GBM TME and localization in tumor niches with special focus on TAM population, its origin and functions in tumor progression and resistance to conventional and experimental GBM treatments. Moreover, recent advances on the development of TAM cell targeting and new cellular therapeutic strategies based on monocyte/macrophages recruitment to eradicate GBM are discussed as complementary therapeutics.
Collapse
|
50
|
Abedin MJ, Michelhaugh SK, Mittal S, Berdichevsky Y. 3D models of glioblastoma interaction with cortical cells. Front Bioeng Biotechnol 2023; 11:1150772. [PMID: 36970613 PMCID: PMC10033518 DOI: 10.3389/fbioe.2023.1150772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction: Glioblastoma (GBM) invasiveness and ability to infiltrate deep into the brain tissue is a major reason for the poor patient prognosis for this type of brain cancer. Behavior of glioblastoma cells, including their motility, and expression of invasion-promoting genes such as matrix metalloprotease-2 (MMP2), are strongly influenced by normal cells found in the brain parenchyma. Cells such as neurons may also be influenced by the tumor, as many glioblastoma patients develop epilepsy. In vitro models of glioblastoma invasiveness are used to supplement animal models in a search for better treatments, and need to combine capability for high-throughput experiments with capturing bidirectional interactions between GBM and brain cells.Methods: In this work, two 3D in vitro models of GBM-cortical interactions were investigated. A matrix-free model was created by co-culturing GBM and cortical spheroids, and a matrix-based model was created by embedding cortical cells and a GBM spheroid in Matrigel.Results: Rapid GBM invasion occurred in the matrix-based model, and was enhanced by the presence of cortical cells. Little invasion occurred in the matrix-free model. In both types of models, presence of GBM cells resulted in a significant increase in paroxysmal neuronal activity.Discussion: Matrix-based model may be better suited for studying GBM invasion in an environment that includes cortical cells, while matrix-free model may be useful in investigation of tumor-associated epilepsy.
Collapse
Affiliation(s)
- Md Joynal Abedin
- Department of Bioengineering, Lehigh University, Bethlehem, PA, United States
| | | | - Sandeep Mittal
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA, United States
| | - Yevgeny Berdichevsky
- Department of Bioengineering, Lehigh University, Bethlehem, PA, United States
- Department of Electrical and Computer Engineering, Bethlehem, PA, United States
- *Correspondence: Yevgeny Berdichevsky,
| |
Collapse
|