1
|
Rajkhowa S, Jha S. The role of NLRP3 and NLRP12 inflammasomes in glioblastoma. Genes Immun 2024; 25:541-551. [PMID: 39604503 DOI: 10.1038/s41435-024-00309-z] [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: 05/07/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is the deadliest malignant brain tumor, with a survival of less than 14 months after diagnosis. The highly invasive nature of GBM makes total surgical resection challenging, leading to tumor recurrence and declined survival. The heterocellular composition of the GBM reprograms its microenvironment, favoring tumor growth, proliferation, and migration. The innate immune cells in the GBM tumor microenvironment, including microglia, astrocytes, and macrophages, express pattern recognition receptors such as NLRs (Nucleotide-binding domain and leucine-rich repeat-containing) that sense pathogen- and damage-associated molecular patterns initiating inflammation. Upon activation, NLRP3 promotes inflammation by NLRP3 inflammasome formation. Auto-proteolytic cleavage and activation of Caspase-1 within the inflammasome leads to caspase-1-mediated cleavage, activation, and conversion of pro-IL-1ß and pro-IL-18 to IL-1ß and IL-18, leading to pyroptosis. In contrast, NLRP12 downregulates inflammatory responses in microglia and macrophages by regulating the NF-κB pathway. NLRP3 and NLRP12 have been implicated in the disease pathophysiology of several cancers with cell-context-dependent, pro- or anti-tumorigenic roles. In this review, we discuss the current literature on the mechanistic roles of NLRP3 and NLRP12 in GBM and the gaps in the scientific literature in the context of GBM pathophysiology with potential for targeted therapeutics.
Collapse
Affiliation(s)
- Sushmita Rajkhowa
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sushmita Jha
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India.
| |
Collapse
|
2
|
Zhao J, Guo P, Fang J, Wang C, Yan C, Bai Y, Wang Z, Du G, Liu A. Neuroinflammation inhibition and neuroprotective effects of purpurin, a potential anti-AD compound, screened via network proximity and gene enrichment analyses. Phytother Res 2024; 38:5474-5489. [PMID: 39351804 DOI: 10.1002/ptr.8064] [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: 06/16/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 10/03/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease without any effective preventive or therapeutic drugs. Natural products with stable structures and pharmacological characteristics are valuable sources for the development of novel drugs for many complex diseases. This study aimed to discover potential natural compounds for the treatment of AD using new technologies and methods and explore the efficacy and mechanism of candidate compounds. AD-related large-scale genetic datasets were collated to construct disease-PPIs and natural products were collected from six databases to construct compound-protein interactions (CPIs). Potential relationships between natural compounds and AD were predicted via network proximity and gene enrichment analyses. Then, five AD-related cell models and d-galactose-induced aging rat model were established to evaluate the neuroprotective effects of candidate compounds in vitro and in vivo. We identified that 267 natural compounds were predicted to have close connections with AD and 19 compounds could exert protective effect in at least one cell model. Notably, purpurin exerted protective effect in three cell models and significantly improved the cognitive learning and memory functions, reduced the oxidative stress injuries and neuroinflammation, and enhanced the synaptic plasticity and neurotrophic effect in the brain of d-galactose-treated rats. In this study, AD-related natural compounds were identified via network proximity and gene enrichment analyses. In vivo and in vitro experiments revealed the therapeutic potential of purpurin for AD treatment, laying the foundation for further in-depth research and providing valuable information for the development of novel anti-AD drugs.
Collapse
Affiliation(s)
- Jun Zhao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pengfei Guo
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chao Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Caiqin Yan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yiming Bai
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhe Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guanhua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ailin Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Lab of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Pizzimenti C, Fiorentino V, Germanò A, Martini M, Ieni A, Tuccari G. Pilocytic astrocytoma: The paradigmatic entity in low‑grade gliomas (Review). Oncol Lett 2024; 27:146. [PMID: 38385109 PMCID: PMC10879958 DOI: 10.3892/ol.2024.14279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/10/2024] [Indexed: 02/23/2024] Open
Abstract
Among low-grade gliomas, representing 10-20% of all primary brain tumours, the paradigmatic entity is constituted by pilocytic astrocytoma (PA), considered a grade 1 tumour by the World Health Organization. Generally, this tumour requires surgical treatment with an infrequent progression towards malignant gliomas. The present review focuses on clinicopathological characteristics, and reports imaging, neurosurgical and molecular features using a multidisciplinary approach. Macroscopically, PA is a slow-growing soft grey tissue, characteristically presenting in association with a cyst and forming a small mural nodule, typically located in the cerebellum, but sometimes occurring in the spinal cord, basal ganglia or cerebral hemisphere. Microscopically, it may appear as densely fibrillated areas composed of elongated pilocytic cells with bipolar 'hairlike' processes or densely fibrillated areas composed of elongated pilocytic cells with Rosenthal fibres alternating with loosely fibrillated areas with a varied degree of myxoid component. A wide range of molecular alterations have been encountered in PA, mostly affecting the MAPK signalling pathway. In detail, the most frequent alteration is a rearrangement of the BRAF gene, although other alterations include neurofibromatosis type-1 mutations, BRAFV600E mutations, KRAS mutations, fibroblast growth factor receptor-1 mutations of fusions, neurotrophic receptor tyrosine kinase family receptor tyrosine kinase fusions and RAF1 gene fusions. The gold standard of PA treatment is surgical excision with complete margin resection, achieving minimal neurological damage. Conventional radiotherapy is not required; the more appropriate treatment appears to be serial follow-up. Chemotherapy should only be applied in younger children to avoid the risk of long-term growth and developmental issues associated with radiation. Finally, if PA recurs, a new surgical approach should be performed. At present, novel therapy involving agents targeting MAPK signalling pathway dysregulation is in development, defining BRAF and MEK inhibitors as target therapeutical agents.
Collapse
Affiliation(s)
- Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, Sections of Pathology and Neurosurgery, University of Messina, I-98125 Messina, Italy
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, Section of Pathology, University of Messina, I-98125 Messina, Italy
| | - Antonino Germanò
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, Sections of Pathology and Neurosurgery, University of Messina, I-98125 Messina, Italy
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, Section of Pathology, University of Messina, I-98125 Messina, Italy
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, Section of Pathology, University of Messina, I-98125 Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age ‘Gaetano Barresi’, Section of Pathology, University of Messina, I-98125 Messina, Italy
| |
Collapse
|
4
|
Wei Y, Li X. Different phenotypes of microglia in animal models of Alzheimer disease. Immun Ageing 2022; 19:44. [PMID: 36209099 PMCID: PMC9547462 DOI: 10.1186/s12979-022-00300-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/19/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022]
Abstract
Microglia are immune-competent cells that are critically involved in maintaining normal brain function. A prominent characteristic of Alzheimer disease (AD) is microglial proliferation and activation concentrated around amyloid plaques in the brain. Recent research has revealed numerous microglial phenotypes related to aging and AD, apart from the traditional M1 and M2 types. Redox signalling modulates the acquisition of the classical or alternative microglia activation phenotypes. The numerous microglial functions can be achieved through these multiple phenotypes, which are associated with distinct molecular signatures.
Collapse
Affiliation(s)
- Yun Wei
- grid.464481.b0000 0004 4687 044XXiyuan Hospital of China Academy of Chinese Medical Sciences, 100091 Beijing, China
| | - Xianxiao Li
- Jingxi Cancer Hospital, 100161 Beijing, China
| |
Collapse
|
5
|
Menna G, Mattogno PP, Donzelli CM, Lisi L, Olivi A, Della Pepa GM. Glioma-Associated Microglia Characterization in the Glioblastoma Microenvironment through a 'Seed-and Soil' Approach: A Systematic Review. Brain Sci 2022; 12:718. [PMID: 35741603 PMCID: PMC9220868 DOI: 10.3390/brainsci12060718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background and aim: Ever since the discovery of tumor-associated immune cells, there has been growing interest in the understanding of the mechanisms underlying the crosstalk between these cells and tumor cells. A "seed and soil" approach has been recently introduced to describe the glioblastoma (GBM) landscape: tumor microenvironments act as fertile "soil" and interact with the "seed" (glial and stem cells compartment). In the following article, we provide a systematic review of the current evidence pertaining to the characterization of glioma-associated macrophages and microglia (GAMs) and microglia and macrophage cells in the glioma tumor microenvironment (TME). Methods: An online literature search was launched on PubMed Medline and Scopus using the following research string: "((Glioma associated macrophages OR GAM OR Microglia) AND (glioblastoma tumor microenvironment OR TME))". The last search for articles pertinent to the topic was conducted in February 2022. Results: The search of the literature yielded a total of 349 results. A total of 235 studies were found to be relevant to our research question and were assessed for eligibility. Upon a full-text review, 58 articles were included in the review. The reviewed papers were further divided into three categories based on their focus: (1) Microglia maintenance of immunological homeostasis and protection against autoimmunity; (2) Microglia crosstalk with dedifferentiated and stem-like glioblastoma cells; (3) Microglia migratory behavior and its activation pattern. Conclusions: Aggressive growth, inevitable recurrence, and scarce response to immunotherapies are driving the necessity to focus on the GBM TME from a different perspective to possibly disentangle its role as a fertile 'soil' for tumor progression and identify within it feasible therapeutic targets. Against this background, our systematic review confirmed microglia to play a paramount role in promoting GBM progression and relapse after treatments. The correct and extensive understanding of microglia-glioma crosstalk could help in understanding the physiopathology of this complex disease, possibly opening scenarios for improvement of treatments.
Collapse
Affiliation(s)
- Grazia Menna
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Pier Paolo Mattogno
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Carlo Maria Donzelli
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Lucia Lisi
- Institute of Pharmacology, Catholic University of Rome, 00168 Rome, Italy;
| | - Alessandro Olivi
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Giuseppe Maria Della Pepa
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| |
Collapse
|
6
|
Characterization of an Immortalized Human Microglial Cell Line as a Tool for the Study of Diabetic Retinopathy. Int J Mol Sci 2022; 23:ijms23105745. [PMID: 35628555 PMCID: PMC9145666 DOI: 10.3390/ijms23105745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 12/04/2022] Open
Abstract
The complexity of the retinal structure reflects on the difficulty to describe its composite cell interactions. Microglia is responsible for the immune reaction to inflammatory stimuli during diabetic retinopathy (DR), but most studies still use rodent cells. We characterized a commercially available immortalized human microglial line and tested its susceptibility to inflammation, to study the interactions between the neuro-vascular retinal portions in species-specific models. After checking the expression of microglial markers, we tried lipopolysaccharide (LPS) stimulation and several pro-inflammatory cocktails to select the best combination able to induce a significant M1 (inflammatory) response. We measured M1 induction through the expression of pro- and anti-inflammatory molecules and performed morphologic and functional assays. Marker expression confirmed the human microglial derivation of these cells. Differently from rodents, LPS did not induce a M1 profile. The best pro-inflammatory stimulus was an interleukin-1β + tumor necrosis factor-α + interferon-γ cocktail, which induced morphology changes and increased proliferation, apoptosis, migration, reactive oxygen species, and the expression of inflammatory cytokines and miRNAs. In conclusion, this microglial line proved potentially useful to investigate the cascade of events leading to DR. In perspective, co-culture models involving microvascular cells will help in the understanding of multifaceted interactions of the neurovascular unit.
Collapse
|
7
|
Niedbała M, Malarz K, Sharma G, Kramer-Marek G, Kaspera W. Glioblastoma: Pitfalls and Opportunities of Immunotherapeutic Combinations. Onco Targets Ther 2022; 15:437-468. [PMID: 35509452 PMCID: PMC9060812 DOI: 10.2147/ott.s215997] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 04/05/2022] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary central nervous system tumour in adults. It has extremely poor prognosis since the current standard of care, comprising of gross total resection and temozolomide (TMZ) chemoradiotherapy, prolongs survival, but does not provide a durable response. To a certain extent, this is due to GBM's heterogeneous, hostile and cold tumour microenvironment (TME) and the unique ability of GBM to overcome the host's immune responses. Therefore, there is an urgent need to develop more effective therapeutic approaches. This review provides critical insights from completed and ongoing clinical studies investigating novel immunotherapy strategies for GBM patients, ranging from the use of immune checkpoint inhibitors in different settings of GBM treatment to novel combinatorial therapies. In particular, we discuss how treatment regimens based on single antigen peptide vaccines evolved into fully personalised, polyvalent cell-based vaccines, CAR-T cell, and viral or gene therapies. Furthermore, the results of the most influential clinical trials and a selection of innovative preclinical studies aimed at activating the immunologically cold GBM microenvironment are reviewed.
Collapse
Affiliation(s)
- Marcin Niedbała
- Department of Neurosurgery, Medical University of Silesia, Regional Hospital, Sosnowiec, Poland
| | - Katarzyna Malarz
- A. Chełkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesia in Katowice, Chorzów, Poland
| | - Gitanjali Sharma
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | | | - Wojciech Kaspera
- Department of Neurosurgery, Medical University of Silesia, Regional Hospital, Sosnowiec, Poland
| |
Collapse
|
8
|
Chang SJ, Chao CT, Kwan AL, Chai CY. The Diagnostic Significance of CXCL13 in M2 Tumor Immune Microenvironment of Human Astrocytoma. Pathol Oncol Res 2022; 28:1610230. [PMID: 35570844 PMCID: PMC9095826 DOI: 10.3389/pore.2022.1610230] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022]
Abstract
Background: CXCL13 may act as a mediator of tumor-associated macrophage immunity during malignant progression. Objective: The present study clarifies the clinicopathological significances of CXCL13 and its corresponding trend with M2 macrophage in human astrocytoma. Methods: The predictive potential of CXCL13 was performed using 695 glioma samples derived from TCGA lower-grade glioma and glioblastoma (GBMLGG) dataset. CXCL13 and M2 biomarker CD163 were observed by immunohistochemistry in 112 astrocytoma tissues. Results: An in-depth analysis showed that CXCL13 expression was related to the poor prognosis of glioma patients (p = 0.0002) derive from TCGA analysis. High level of CXCL13 was detected in 43 (38.39%) astrocytoma and CXCL13/CD163 coexpression was expressed in 33 (29.46%) cases. The immunoreactivities of CXCL13 and CXCL13/CD163 were found in the malignant lesions, which were both significantly associated with grade, patient survival, and IDH1 mutation. Single CXCL13 and CXCL13/CD163 coexpression predicted poor overall survival in astrocytoma (p = 0.0039 and p = 0.0002, respectively). Multivariate Cox regression analyses manifested CXCL13/CD163 phenotype was a significant independent prognostic indicator of patient outcome in astrocytoma (CXCL13, p = 0.0642; CXCL13/CD163, p = 0.0368). Conclusion: CXCL13 overexpression is strongly linked to CD163+ M2 infiltration in malignant astrocytoma. CXCL13/CD163 coexpression would imply M2c-related aggressive characteristics existing in astrocytoma progression could also provide predictive trends of patient outcomes.
Collapse
Affiliation(s)
- Shu-Jyuan Chang
- Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chia-Te Chao
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
9
|
Tamai S, Ichinose T, Tsutsui T, Tanaka S, Garaeva F, Sabit H, Nakada M. Tumor Microenvironment in Glioma Invasion. Brain Sci 2022; 12:brainsci12040505. [PMID: 35448036 PMCID: PMC9031400 DOI: 10.3390/brainsci12040505] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/05/2023] Open
Abstract
A major malignant trait of gliomas is their remarkable infiltration capacity. When glioma develops, the tumor cells have already reached the distant part. Therefore, complete removal of the glioma is impossible. Recently, research on the involvement of the tumor microenvironment in glioma invasion has advanced. Local hypoxia triggers cell migration as an environmental factor. The transcription factor hypoxia-inducible factor (HIF) -1α, produced in tumor cells under hypoxia, promotes the transcription of various invasion related molecules. The extracellular matrix surrounding tumors is degraded by proteases secreted by tumor cells and simultaneously replaced by an extracellular matrix that promotes infiltration. Astrocytes and microglia become tumor-associated astrocytes and glioma-associated macrophages/microglia, respectively, in relation to tumor cells. These cells also promote glioma invasion. Interactions between glioma cells actively promote infiltration of each other. Surgery, chemotherapy, and radiation therapy transform the microenvironment, allowing glioma cells to invade. These findings indicate that the tumor microenvironment may be a target for glioma invasion. On the other hand, because the living body actively promotes tumor infiltration in response to the tumor, it is necessary to reconsider whether the invasion itself is friend or foe to the brain.
Collapse
|
10
|
Tu S, Lin X, Qiu J, Zhou J, Wang H, Hu S, Yao Y, Wang Y, Deng Y, Zhou Y, Shao A. Crosstalk Between Tumor-Associated Microglia/Macrophages and CD8-Positive T Cells Plays a Key Role in Glioblastoma. Front Immunol 2021; 12:650105. [PMID: 34394072 PMCID: PMC8358794 DOI: 10.3389/fimmu.2021.650105] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/21/2021] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma is considered to be the most malignant disease of the central nervous system, and it is often associated with poor survival. The immune microenvironment plays a key role in the development and treatment of glioblastoma. Among the different types of immune cells, tumor-associated microglia/macrophages (TAM/Ms) and CD8-positive (CD8+) T cells are the predominant immune cells, as well as the most active ones. Current studies have suggested that interaction between TAM/Ms and CD8+ T cells have numerous potential targets that will allow them to overcome malignancy in glioblastoma. In this review, we summarize the mechanism and function of TAM/Ms and CD8+ T cells involved in glioblastoma, as well as update on the relationship and crosstalk between these two cell types, to determine whether this association alters the immune status during glioblastoma development and affects optimal treatment. We focus on the molecular factors that are crucial to this interaction, and the role that this crosstalk plays in the biological processes underlying glioblastoma treatment, particularly with regard to immune therapy. We also discuss novel therapeutic targets that can aid in resolving reticular connections between TAM/Ms and CD8+ T cells, including depletion and reprogramming TAM/Ms and novel TAM/Ms-CD8+ T cell cofactors with potential translational usage. In addition, we highlight the challenges and discuss future perspectives of this crosstalk between TAM/Ms and CD8+ T cells.
Collapse
Affiliation(s)
- Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xu Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jili Qiu
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaqi Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Wang
- Department of Medical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiyao Hu
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Yihan Yao
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Comparison of In Vivo and Ex Vivo Magnetic Resonance Imaging in a Rat Model for Glioblastoma-Associated Epilepsy. Diagnostics (Basel) 2021; 11:diagnostics11081311. [PMID: 34441246 PMCID: PMC8393600 DOI: 10.3390/diagnostics11081311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 11/17/2022] Open
Abstract
Magnetic resonance imaging (MRI) is frequently used for preclinical treatment monitoring in glioblastoma (GB). Discriminating between tumors and tumor-associated changes is challenging on in vivo MRI. In this study, we compared in vivo MRI scans with ex vivo MRI and histology to estimate more precisely the abnormal mass on in vivo MRI. Epileptic seizures are a common symptom in GB. Therefore, we used a recently developed GB-associated epilepsy model from our group with the aim of further characterizing the model and making it useful for dedicated epilepsy research. Ten days after GB inoculation in rat entorhinal cortices, in vivo MRI (T2w and mean diffusivity (MD)), ex vivo MRI (T2w) and histology were performed, and tumor volumes were determined on the different modalities. The estimated abnormal mass on ex vivo T2w images was significantly smaller compared to in vivo T2w images, but was more comparable to histological tumor volumes, and might be used to estimate end-stage tumor volumes. In vivo MD images displayed tumors as an outer rim of hyperintense signal with a core of hypointense signal, probably reflecting peritumoral edema and tumor mass, respectively, and might be used in the future to distinguish the tumor mass from peritumoral edema—associated with reactive astrocytes and activated microglia, as indicated by an increased expression of immunohistochemical markers—in preclinical models. In conclusion, this study shows that combining imaging techniques using different structural scales can improve our understanding of the pathophysiology in GB.
Collapse
|
12
|
Zhang B, Zhao J, Wang Z, Guo P, Liu A, Du G. Identification of Multi-Target Anti-AD Chemical Constituents From Traditional Chinese Medicine Formulae by Integrating Virtual Screening and In Vitro Validation. Front Pharmacol 2021; 12:709607. [PMID: 34335272 PMCID: PMC8322649 DOI: 10.3389/fphar.2021.709607] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that seriously threatens the health of the elderly. At present, no drugs have been proven to cure or delay the progression of the disease. Due to the multifactorial aetiology of this disease, the multi-target-directed ligand (MTDL) approach provides an innovative and promising idea in search for new drugs against AD. In order to find potential multi-target anti-AD drugs from traditional Chinese medicine (TCM) formulae, a compound database derived from anti-AD Chinese herbal formulae was constructed and predicted by the anti-AD multi-target drug prediction platform established in our laboratory. By analyzing the results of virtual screening, 226 chemical constituents with 3 or more potential AD-related targets were collected, from which 16 compounds that were predicted to combat AD through various mechanisms were chosen for biological validation. Several cell models were established to validate the anti-AD effects of these compounds, including KCl, Aβ, okadaic acid (OA), SNP and H2O2 induced SH-SY5Y cell model and LPS induced BV2 microglia model. The experimental results showed that 12 compounds including Nonivamide, Bavachromene and 3,4-Dimethoxycinnamic acid could protect model cells from AD-related damages and showed potential anti-AD activity. Furthermore, the potential targets of Nonivamide were investigated by molecular docking study and analysis with CDOCKER revealed the possible binding mode of Nonivamide with its predicted targets. In summary, 12 potential multi-target anti-AD compounds have been found from anti-AD TCM formulae by comprehensive application of computational prediction, molecular docking method and biological validation, which laid a theoretical and experimental foundation for in-depth study, also providing important information and new research ideas for the discovery of anti-AD compounds from traditional Chinese medicine.
Collapse
Affiliation(s)
- Baoyue Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zhao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Wang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengfei Guo
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ailin Liu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guanhua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Ma J, Chen CC, Li M. Macrophages/Microglia in the Glioblastoma Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22115775. [PMID: 34071306 PMCID: PMC8198046 DOI: 10.3390/ijms22115775] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 12/23/2022] Open
Abstract
The complex interaction between glioblastoma and its microenvironment has been recognized for decades. Among various immune profiles, the major population is tumor-associated macrophage, with microglia as its localized homolog. The present definition of such myeloid cells is based on a series of cell markers. These good sentinel cells experience significant changes, facilitating glioblastoma development and protecting it from therapeutic treatments. Huge, complicated mechanisms are involved during the overall processes. A lot of effort has been dedicated to crack the mysterious codes in macrophage/microglia recruiting, activating, reprogramming, and functioning. We have made our path. With more and more key factors identified, a lot of new therapeutic methods could be explored to break the ominous loop, to enhance tumor sensitivity to treatments, and to improve the prognosis of glioblastoma patients. However, it might be a synergistic system rather than a series of clear, stepwise events. There are still significant challenges before the light of truth can shine onto the field. Here, we summarize recent advances in this field, reviewing the path we have been on and where we are now.
Collapse
Affiliation(s)
| | | | - Ming Li
- Correspondence: (C.C.C.); (M.L.)
| |
Collapse
|
14
|
Jin Y, Kang Y, Peng X, Yang L, Li Q, Mei Q, Chen X, Hu G, Tang Y, Yuan X. Irradiation-Induced Activated Microglia Affect Brain Metastatic Colonization of NSCLC Cells via miR-9/ CDH1 Axis. Onco Targets Ther 2021; 14:1911-1922. [PMID: 33758511 PMCID: PMC7981147 DOI: 10.2147/ott.s301412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Brain metastasis is among the leading causes of death in patients with non-small-cell lung cancer (NSCLC). Through yet unknown mechanisms, prophylactic cranial irradiation (PCI) can significantly decrease the incidence of brain metastases. Given that PCI probably exerts indirect anti-tumoral effects by turning cerebral "soil" unfavorable for the colonization of metastatic tumor "seeds". This study aims to reveal how PCI regulates the brain microenvironment conducing to a reduction in brain metastases. MATERIALS AND METHODS Key markers of M1/M2 microglia types and mesenchymal-to-epithelial transition (MET) were analyzed by qRT-PCR and Western Blot in vitro. The target miR-9 was obtained by miRNA array analysis and confirmed by qRT-PCR in microglia. We used miRTarBase and TargetScan to analyze the target genes of miR-9 and confirmed by luciferase activity assay. Anti-metastatic effects of irradiation on the brain were evaluated by intravital imaging using a brain metastatic A549-F3 cell line in a nude mouse model. RESULTS Irradiation induced M1 microglia activation, which inhibited the MET process of A549 cell lines. Furthermore, levels of miR-9 secreted by irradiated M1 microglia significantly increased and played a vital role in the inhibition of the A549 MET process by directly targeting CDH1, concurrently decreasing cell capacity for localization in the brain, thus reducing brain metastases. CONCLUSION We demonstrated that miR-9 secreted by irradiated M1-type microglia played an important role in modulating A549 cell lines into mesenchymal phenotype and further decreased their localization capabilities in the brain. Our findings signify the modulating effect of irradiation on metastatic soil and the cross-talk between tumour cells and the metastatic microenvironment; importantly, they provide new opportunities for effective anti-metastasis therapies, especially for brain metastasis patients.
Collapse
Affiliation(s)
- Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Yalin Kang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Xiaohong Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Li Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Yang Tang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
15
|
Zhuang L, Kong Y, Yang S, Lu F, Gong Z, Zhan S, Liu M. Dynamic changes of inflammation and apoptosis in cerebral ischemia‑reperfusion injury in mice investigated by ferumoxytol‑enhanced magnetic resonance imaging. Mol Med Rep 2021; 23:282. [PMID: 33604682 PMCID: PMC7905325 DOI: 10.3892/mmr.2021.11921] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 03/13/2020] [Indexed: 01/21/2023] Open
Abstract
The inflammatory response and apoptosis are key factors in cerebral ischemia-reperfusion injury. The severity of the inflammatory reaction and apoptosis has an important impact on the prognosis of stroke. The ultrasmall superparamagnetic iron oxide particle has provided an effective magnetic resonance molecular imaging method for dynamic observation of the cell infiltration process in vivo. The aims of the present study were to investigate the inflammatory response of cerebral ischemia-reperfusion injury in mice using ferumoxytol-enhanced magnetic resonance imaging, and to observe the dynamic changes of inflammatory response and apoptosis. In the present study a C57BL/6n mouse cerebral ischemia-reperfusion model was established by blocking the right middle cerebral artery with an occluding suture. Subsequently, the mice were injected with ferumoxytol via the tail vein, and magnetic resonance scanning was performed at corresponding time points to observe the signal changes. Furthermore, blood samples were used to measure the level of serum inflammatory factors, and histological staining was performed to assess the number of iron-swallowing microglial cells and apoptotic cells. The present results suggested that there was no significant difference in the serum inflammatory factors tumor necrosis factor-α and interleukin 1β between the middle cerebral artery occlusion (MCAO) and MCAO + ferumoxytol groups injected with ferumoxytol and physiological saline. The lowest signal ratio in the negative enhancement region was decreased 24 h after reperfusion in mice injected with ferumoxytol. The proportion of iron-swallowing microglial cells and TUNEL-positive cells were the highest at 24 h after reperfusion, and decreased gradually at 48 and 72 h after reperfusion. Therefore, the present results indicated that ferumoxytol injection of 18 mg Fe/kg does not affect the inflammatory response in the acute phase of cerebral ischemia and reperfusion. Ferumoxytol-enhanced magnetic resonance imaging can be used as an effective means to monitor the inflammatory response in the acute phase of cerebral ischemia-reperfusion injury. Furthermore, it was found that activation of the inflammatory response and apoptosis in the acute stage of cerebral ischemia-reperfusion injury is consistent.
Collapse
Affiliation(s)
- Lihua Zhuang
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yingnan Kong
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Shuohui Yang
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Fang Lu
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Zhigang Gong
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Songhua Zhan
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Mengxiao Liu
- MR Scientific Marketing, Siemens Healthcare, Shanghai 201318, P.R. China
| |
Collapse
|
16
|
Hasan MN, Luo L, Ding D, Song S, Bhuiyan MIH, Liu R, Foley LM, Guan X, Kohanbash G, Hitchens TK, Castro MG, Zhang Z, Sun D. Blocking NHE1 stimulates glioma tumor immunity by restoring OXPHOS function of myeloid cells. Theranostics 2021; 11:1295-1309. [PMID: 33391535 PMCID: PMC7738877 DOI: 10.7150/thno.50150] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Immunosuppressive tumor microenvironment (TME) in glioblastoma (GBM) is one of the contributing factors for failed immunotherapies. Therefore, there is an urgent need to better understand TME and to identify novel modulators of TME for more effective GBM therapies. We hypothesized that H+ extrusion protein Na/H exchanger 1 (NHE1) plays a role in dysregulation of glucose metabolism and immunosuppression of GBM. We investigated the efficacy of blockade of NHE1 activity in combination with temozolomide (TMZ) therapy in increasing anti-tumor immunity. Methods: Mouse syngeneic intracranial glioma model was used to test four treatment regimens: DMSO (Vehicle-control), TMZ, NHE1 specific inhibitor HOE642, or TMZ+HOE642 (T+H) combination. Ex vivo 1H/19Fluorine magnetic resonance imaging (MRI) with cell tracking agent Vsense was performed to monitor the infiltration of glioma-associated microglia/myeloid cells (GAMs). Glucose metabolism and transcriptome profiles were analyzed by Seahorse analyzer and bulk RNA-sequencing. The impact of selective Nhe1 deletion in GAMs on sensitivity to anti-PD-1 therapy was evaluated in transgenic NHE1 knockout (KO) mice. Results: Among the tested treatment regimens, the T+H combination therapy significantly stimulated the infiltration of GAMs and T-cells; up-regulated Th1 activation, and mitochondrial oxidative phosphorylation (OXPHOS) pathway genes, increased glucose uptake and mitochondrial mass, and decreased aerobic glycolysis in GAMs. Selective deletion of Nhe1 in Cx3cr1+ Nhe1 KO mice increased anti-tumor immunity and sensitivity to TMZ plus anti-PD-1 combinatorial therapy. Conclusions: NHE1 plays a role in developing glioma immunosuppressive TME in part by dysregulating glucose metabolism of GAMs and emerges as a therapeutic target for improving glioma immunity.
Collapse
|
17
|
Martins TA, Schmassmann P, Shekarian T, Boulay JL, Ritz MF, Zanganeh S, Vom Berg J, Hutter G. Microglia-Centered Combinatorial Strategies Against Glioblastoma. Front Immunol 2020; 11:571951. [PMID: 33117364 PMCID: PMC7552736 DOI: 10.3389/fimmu.2020.571951] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated microglia (MG) and macrophages (MΦ) are important components of the glioblastoma (GBM) immune tumor microenvironment (iTME). From the recent advances in understanding how MG and GBM cells evolve and interact during tumorigenesis, we emphasize the cooperation of MG with other immune cell types of the GBM-iTME, mainly MΦ and T cells. We provide a comprehensive overview of current immunotherapeutic clinical trials and approaches for the treatment of GBM, which in general, underestimate the counteracting contribution of immunosuppressive MG as a main factor for treatment failure. Furthermore, we summarize new developments and strategies in MG reprogramming/re-education in the GBM context, with a focus on ways to boost MG-mediated tumor cell phagocytosis and associated experimental models and methods. This ultimately converges in our proposal of novel combinatorial regimens that locally modulate MG as a central paradigm, and therefore may lead to additional, long-lasting, and effective tumoricidal responses.
Collapse
Affiliation(s)
- Tomás A Martins
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Tala Shekarian
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jean-Louis Boulay
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Marie-Françoise Ritz
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Steven Zanganeh
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Chemical and Biomolecular Engineering, New York University, New York, NY, United States
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
| | - Gregor Hutter
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
18
|
Shono K, Yamaguchi I, Mizobuchi Y, Kagusa H, Sumi A, Fujihara T, Nakajima K, Kitazato KT, Matsuzaki K, Saya H, Takagi Y. Downregulation of the CCL2/CCR2 and CXCL10/CXCR3 axes contributes to antitumor effects in a mouse model of malignant glioma. Sci Rep 2020; 10:15286. [PMID: 32943658 PMCID: PMC7499211 DOI: 10.1038/s41598-020-71857-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 08/17/2020] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme involves glioma stem cells (GSCs) that are resistant to various therapeutic approaches. Here, we studied the importance of paracrine signaling in the glioma microenvironment by focusing on the celecoxib-mediated role of chemokines C–C motif ligand 2 (CCL2), C-X-C ligand 10 (CXCL10), and their receptors, CCR2 and CXCR3, in GSCs and a GSC-bearing malignant glioma model. C57BL/6 mice were injected with orthotopic GSCs intracranially and divided into groups administered either 10 or 30 mg/kg celecoxib, or saline to examine the antitumor effects associated with chemokine expression. In GSCs, we analyzed cell viability and expression of chemokines and their receptors in the presence/absence of celecoxib. In the malignant glioma model, celecoxib exhibited antitumor effects in a dose dependent manner and decreased protein and mRNA levels of Ccl2 and CxcL10 and Cxcr3 but not of Ccr2. CCL2 and CXCL10 co-localized with Nestin+ stem cells, CD16+ or CD163+ macrophages and Iba-1+ microglia. In GSCs, celecoxib inhibited Ccl2 and Cxcr3 expression in a nuclear factor-kappa B-dependent manner but not Ccr2 and CxcL10. Moreover, Ccl2 silencing resulted in decreased GSC viability. These results suggest that celecoxib-mediated regulation of the CCL2/CCR2 and CXCL10/ CXCR3 axes may partially contribute to glioma-specific antitumor effects.
Collapse
Affiliation(s)
- Kenji Shono
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Izumi Yamaguchi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yoshifumi Mizobuchi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan.
| | - Hiroshi Kagusa
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Akiko Sumi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Toshitaka Fujihara
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Kohei Nakajima
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Keiko T Kitazato
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Kazuhito Matsuzaki
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yasushi Takagi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| |
Collapse
|
19
|
Afshari AR, Mollazadeh H, Sahebkar A. Minocycline in Treating Glioblastoma Multiforme: Far beyond a Conventional Antibiotic. JOURNAL OF ONCOLOGY 2020; 2020:8659802. [PMID: 33014057 PMCID: PMC7519463 DOI: 10.1155/2020/8659802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/05/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
Abstract
One of the most lethal forms of CNS pathologies is glioblastoma multiforme (GBM) that represents high invasiveness, uncontrolled proliferation, and angiogenic features. Its invasiveness is responsible for the high recurrence even after maximal surgical interventions. Minocycline is a semisynthetic analog of tetracyclines with potential anti-inflammatory and anticancer effects, distinct from its antimicrobial activity. In this review, we highlight the importance and the cytotoxic mechanisms of minocycline on GBM pathophysiology. Considering the role of certain enzymes in autophagy, apoptosis, tumor cell invasion, and metastatic ability, the possible use of tetracyclines for cancer therapy should be investigated, especially GBM. The present study is, therefore, going to cover the main topics in minocycline pharmacology to date, encouraging its consideration as a new treatment approach for cancer and GBM.
Collapse
Affiliation(s)
- Amir R. Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Mellai M, Annovazzi L, Bisogno I, Corona C, Crociara P, Iulini B, Cassoni P, Casalone C, Boldorini R, Schiffer D. Chondroitin Sulphate Proteoglycan 4 (NG2/CSPG4) Localization in Low- and High-Grade Gliomas. Cells 2020; 9:E1538. [PMID: 32599896 PMCID: PMC7349878 DOI: 10.3390/cells9061538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Neuron glial antigen 2 or chondroitin sulphate proteoglycan 4 (NG2/CSPG4) is expressed by immature precursors/progenitor cells and is possibly involved in malignant cell transformation. The aim of this study was to investigate its role on the progression and survival of sixty-one adult gliomas and nine glioblastoma (GB)-derived cell lines. METHODS NG2/CSPG4 protein expression was assessed by immunohistochemistry and immunofluorescence. Genetic and epigenetic alterations were detected by molecular genetic techniques. RESULTS NG2/CSPG4 was frequently expressed in IDH-mutant/1p19q-codel oligodendrogliomas (59.1%) and IDH-wild type GBs (40%) and rarely expressed in IDH-mutant or IDH-wild type astrocytomas (14.3%). Besides tumor cells, NG2/CSPG4 immunoreactivity was found in the cytoplasm and/or cell membranes of reactive astrocytes and vascular pericytes/endothelial cells. In GB-derived neurospheres, it was variably detected according to the number of passages of the in vitro culture. In GB-derived adherent cells, a diffuse positivity was found in most cells. NG2/CSPG4 expression was significantly associated with EGFR gene amplification (p = 0.0005) and poor prognosis (p = 0.016) in astrocytic tumors. CONCLUSION The immunoreactivity of NG2/CSPG4 provides information on the timing of the neoplastic transformation and could have prognostic and therapeutic relevance as a promising tumor-associated antigen for antibody-based immunotherapy in patients with malignant gliomas.
Collapse
Affiliation(s)
- Marta Mellai
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100 Novara, Italy; (M.M.); (R.B.)
- Centro Interdipartimentale di Ricerca Traslazionale sulle Malattie Autoimmuni e Allergiche (CAAD), Università del Piemonte Orientale (UPO), Corso Trieste 15A, 28100 Novara, Italy
- Fondazione Edo ed Elvo Tempia Valenta—ONLUS, Via Malta 3, 13900 Biella, Italy
| | - Laura Annovazzi
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| | - Ilaria Bisogno
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| | - Cristiano Corona
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Paola Crociara
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Barbara Iulini
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Paola Cassoni
- Dipartimento di Scienze Mediche, Università di Torino/Città della Salute e della Scienza, Via Santena 7, 10126 Torino, Italy;
| | - Cristina Casalone
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Renzo Boldorini
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100 Novara, Italy; (M.M.); (R.B.)
| | - Davide Schiffer
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| |
Collapse
|
21
|
Marinari E, Allard M, Gustave R, Widmer V, Philippin G, Merkler D, Tsantoulis P, Dutoit V, Dietrich PY. Inflammation and lymphocyte infiltration are associated with shorter survival in patients with high-grade glioma. Oncoimmunology 2020; 9:1779990. [PMID: 32923142 PMCID: PMC7458651 DOI: 10.1080/2162402x.2020.1779990] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Glioma represents a serious health burden in terms of morbidity and mortality. The prognostic significance of the lymphoid and myeloid infiltrates in glioma is not clearly determined. Moreover, the characterization of different leukocyte subsets in the tumor microenvironment relies mainly on immunohistochemistry observations, and data about their association with prognosis are contradictory. Here, we performed acomprehensive study of both the tumor-infiltrating and circulating immune compartments of patients with high-grade glioma. Nineteen tumor biopsies and 30 PBMC samples were analyzed by RNA sequencing. Validation was performed on The Cancer Genome Atlas (TCGA) RNA sequencing data from glioma and on additional 39 tumor biopsies analyzed by flow cytometry. We identified prognostic tumor and peripheral immune signatures, which associate increased inflammation, immune infiltration and activation with shorter overall survival in high-grade glioma patients. Importantly, we confirmed our observations by flow cytometry analysis and validated the tumor-signature using the TCGA dataset. In addition, both tumor genotype and grade associated with the degree of glioma immune infiltration. Unlike in the majority of cancers, lymphocyte infiltration at the tumor site is anegative prognostic factor in glioma, suggesting the ambivalent pro-tumorigenic role of immune responses in glioma.
Collapse
Affiliation(s)
- Eliana Marinari
- Laboratory of Tumor Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Mathilde Allard
- Laboratory of Tumor Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Robin Gustave
- Laboratory of Tumor Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Valérie Widmer
- Laboratory of Tumor Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Géraldine Philippin
- Laboratory of Tumor Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Petros Tsantoulis
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Oncology Service, Geneva University Hospitals, Geneva, Switzerland
| | - Valérie Dutoit
- Laboratory of Tumor Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Oncology Service, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
22
|
Chang SJ, Tu HP, Lai YCC, Luo CW, Nejo T, Tanaka S, Chai CY, Kwan AL. Increased Vascular Adhesion Protein 1 (VAP-1) Levels are Associated with Alternative M2 Macrophage Activation and Poor Prognosis for Human Gliomas. Diagnostics (Basel) 2020; 10:diagnostics10050256. [PMID: 32349342 PMCID: PMC7278017 DOI: 10.3390/diagnostics10050256] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 12/19/2022] Open
Abstract
Glioma is characterized by a high heterogeneity in the brain tumor. Abundant tumor-associated macrophages (TAMs) exist as neoplastic tissues, implicating tumor plasticity and thus leading to therapeutic challenges. Vascular adhesion protein (VAP-1) potentially serves as a mediator for TAM immunity in tumor milieu. We previously demonstrated that VAP-1 could contribute to tumor malignancy, but its characteristics in TAM immunity of glioma progression are still unclear. This study explored the association of VAP-1 expression with TAM distribution as well as the resulting clinical significance and prognostic value in human gliomas. An in-depth analysis of AOC3 (VAP-1) gene expression was performed using 695 glioma samples derived from the cancer genome atlas (TCGA)-lower grade glioma and glioblastoma (GBMLGG) cohort. Bioinformatic analysis confirmed that VAP-1 expression is associated with poor prognosis of glioma patients (p = 0.0283). VAP-1 and TAM biomarkers (CD68, iNOS, and CD163) were evaluated by immunohistochemistry in 108 gliomas from Kaohsiung Medical University Hospital. VAP-1+ was expressed in 56 (51.85%) cases and this phenotype revealed a significant association with overall survival in Kaplan–Meier analysis (p < 0.0001). Immunohistochemical double staining showed that VAP-1 immunoreactivity was present around CD163+ M2 infiltration location, including aggressive lesions and neighboring neovasculature. We demonstrated that high VAP-1 expression levels positively correlated with CD163+ M2 activation and coexpression of these two proteins was associated with worse survival in gliomas (p < 0.0001). Multivariate analysis indicated that VAP-1 alone and co-expressed with CD163 were the significantly independent indicators (both p < 0.0001). Furthermore, VAP-1/CD163 coexpression exhibited excellent diagnostic accuracy in gliomas (AUC = 0.8008). In conclusion, VAP-1 and TAM CD163 M2 coexpression was found in glioma tissues belonging to a highly malignant subgroup that was associated with poor prognosis. These results implied VAP-1 abundance is closely linked to alternative M2 activation during glioma progression. From the aforementioned data, a reasonable inference is that VAP-1 combined with targeting M2 immunity might be an effective therapeutic target for human gliomas.
Collapse
Affiliation(s)
- Shu-Jyuan Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Hung-Pin Tu
- Department of Public Health and Environmental Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yen-Chang Clark Lai
- Department of Pathology, Kaohsiung Medical University Chung Ho Memorial Hospital, Kaohsiung 80756, Taiwan;
| | - Chi-Wen Luo
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University Chung Ho Memorial Hospital, Kaohsiung 80756, Taiwan;
- Department of Surgery, Kaohsiung Medical University Chung Ho Memorial Hospital, Kaohsiung 80756, Taiwan
| | - Takahide Nejo
- Department of Neurosurgery, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan; (T.N.); (S.T.)
| | - Shota Tanaka
- Department of Neurosurgery, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan; (T.N.); (S.T.)
| | - Chee-Yin Chai
- Department of Pathology, Kaohsiung Medical University Chung Ho Memorial Hospital, Kaohsiung 80756, Taiwan;
- Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Correspondence: (C.-Y.C.); (A.-L.K.); Tel.: +88-6-7312-1101 (ext. 7081) (C.-Y.C.); +88-6-7312-1101 (ext. 5880) (A.-L.K.); Fax: +88-6-7313-6681 (C.-Y.C.); +88-6-7321-5039 (A.-L.K.)
| | - Aij-Lie Kwan
- Department of Neurosurgery, Kaohsiung Medical University Chung Ho Memorial Hospital, Kaohsiung 80756, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (C.-Y.C.); (A.-L.K.); Tel.: +88-6-7312-1101 (ext. 7081) (C.-Y.C.); +88-6-7312-1101 (ext. 5880) (A.-L.K.); Fax: +88-6-7313-6681 (C.-Y.C.); +88-6-7321-5039 (A.-L.K.)
| |
Collapse
|
23
|
Gheorghe RO, Deftu A, Filippi A, Grosu A, Bica-Popi M, Chiritoiu M, Chiritoiu G, Munteanu C, Silvestro L, Ristoiu V. Silencing the Cytoskeleton Protein Iba1 (Ionized Calcium Binding Adapter Protein 1) Interferes with BV2 Microglia Functioning. Cell Mol Neurobiol 2020; 40:1011-1027. [PMID: 31950314 DOI: 10.1007/s10571-020-00790-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/07/2020] [Indexed: 01/31/2023]
Abstract
Iba1 (ionized calcium binding adapter protein 1) is a cytoskeleton protein specific only for microglia and macrophages, where it acts as an actin-cross linking protein. Although frequently regarded as a marker of activation, its involvement in cell migration, membrane ruffling, phagocytosis or in microglia remodeling during immunological surveillance of the brain suggest that Iba1 is not a simple cytoskeleton protein, but a signaling molecule involved in specific signaling pathways. In this study we investigated if Iba1 could also represent a drug target, and tested the hypothesis that its specific silencing with customized Iba1-siRNA can modulate microglia functioning. The results showed that Iba1-silenced BV2 microglia migrate less due to reduced proliferation and cell adhesion, while their phagocytic activity and P2x7 functioning was significantly increased. Our data are the proof of concept that Iba1 protein is a new microglia target, which opens a new therapeutic avenue for modulating microglia behavior.
Collapse
Affiliation(s)
- Roxana-Olimpia Gheorghe
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Alexandru Deftu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Alexandru Filippi
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Andreea Grosu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Melania Bica-Popi
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Marioara Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Gabriela Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Cristian Munteanu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Luigi Silvestro
- Pharma Serv International, Sabinelor 52, 050853, Bucharest, Romania
| | - Violeta Ristoiu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania.
| |
Collapse
|
24
|
Role of Infiltrating Microglia/Macrophages in Glioma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:281-298. [PMID: 32034719 DOI: 10.1007/978-3-030-30651-9_14] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this chapter we describe the state of the art knowledge of the role played by myeloid cells in promoting and supporting the growth and the invasive properties of a deadly brain tumor, glioblastoma. We provide a review of the works describing the intercellular communication among glioma and associated microglia/macrophage cells (GAMs) using in vitro cellular models derived from mice, rats and human patients and in vivo animal models using syngeneic or xenogeneic experimental systems. Special emphasis will be given to 1) the timing alteration of brain microenvironment under the influence of glioma, 2) the bidirectional communication among tumor and GAMs, 3) possible approaches to interfere with or to guide these interactions, with the aim to identify molecular and cellular targets which could revert or delay the vicious cycle that favors tumor biology.
Collapse
|
25
|
Bhavya B, Anand CR, Madhusoodanan UK, Rajalakshmi P, Krishnakumar K, Easwer HV, Deepti AN, Gopala S. To be Wild or Mutant: Role of Isocitrate Dehydrogenase 1 (IDH1) and 2-Hydroxy Glutarate (2-HG) in Gliomagenesis and Treatment Outcome in Glioma. Cell Mol Neurobiol 2020; 40:53-63. [PMID: 31485826 DOI: 10.1007/s10571-019-00730-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022]
Abstract
Molecular and clinical research based on isocitrate dehydrogenase (IDH) mutations is much sought after in glioma research since a decade of its discovery in 2008. IDH enzyme normally catalyzes isocitrate to α-keto-glutarate (α-KG), but once the gene is mutated it produces an 'oncometabolite', 2-hydroxyglutarate (2-HG). 2-HG is proposed to inhibit α-KG-dependent dioxygenases and also blocks cellular differentiation. Here, we discuss the role of the IDH1 mutation in gliomagenesis. The review also focuses on the effect of 2-HG on glioma epigenetics, the cellular signaling involved in IDH1 mutant glioma cells and the therapeutic response seen in mutant IDH1(mIDH1) harboring glioma patients in comparison to the patients with wild-type IDH1. The review encompasses the debatable impacts of the mutation on immune microenvironment a propos of various mIDH1 inhibitors in practice or in trials. Recent studies revealing the relation of IDH mutation with the immune microenvironment and inflammatory status in untreated versus treated glioblastoma patients are highlighted with respect to prospective therapeutic targets. Also at the molecular level, the association of mIDH1/2-HG with the intracellular components such as mitochondria and other neighboring cells is discussed.
Collapse
Affiliation(s)
- Bharathan Bhavya
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - C R Anand
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - U K Madhusoodanan
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - P Rajalakshmi
- Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - K Krishnakumar
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - H V Easwer
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - A N Deepti
- Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695011, India.
| |
Collapse
|
26
|
Yamaguchi I, Nakajima K, Shono K, Mizobuchi Y, Fujihara T, Shikata E, Yamaguchi T, Kitazato K, Sampetrean O, Saya H, Takagi Y. Downregulation of PD-L1 via FKBP5 by celecoxib augments antitumor effects of PD-1 blockade in a malignant glioma model. Neurooncol Adv 2019; 2:vdz058. [PMID: 32642723 PMCID: PMC7212915 DOI: 10.1093/noajnl/vdz058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Antitumor therapies targeting programmed cell death-1 (PD-1) or its ligand-1 (PD-L1) are used in various cancers. However, in glioblastoma (GBM), the expression of PD-L1 varies between patients, and the relationship between this variation and the efficacy of anti-PD-1 antibody therapy remains unclear. High expression levels of PD-L1 affect the proliferation and invasiveness of GBM cells. As COX-2 modulates PD-L1 expression in cancer cells, we tested the hypothesis that the COX-2 inhibitor celecoxib potentiates anti-PD-1 antibody treatment via the downregulation of PD-L1. Methods Six-week-old male C57BL/6 mice injected with murine glioma stem cells (GSCs) were randomly divided into four groups treated with vehicle, celecoxib, anti-PD-1 antibody, or celecoxib plus anti-PD-1 antibody and the antitumor effects of these treatments were assessed. To verify the mechanisms underlying these effects, murine GSCs and human GBM cells were studied in vitro. Results Compared with that with each single treatment, the combination of celecoxib and anti-PD-1 antibody treatment significantly decreased tumor volume and prolonged survival. The high expression of PD-L1 was decreased by celecoxib in the glioma model injected with murine GSCs, cultured murine GSCs, and cultured human GBM cells. This reduction was associated with post-transcriptional regulation of the co-chaperone FK506-binding protein 5 (FKBP5). Conclusions Combination therapy with anti-PD-1 antibody plus celecoxib might be a promising therapeutic strategy to target PD-L1 in glioblastoma. The downregulation of highly-expressed PD-L1 via FKBP5, induced by celecoxib, could play a role in its antitumor effects.
Collapse
Affiliation(s)
- Izumi Yamaguchi
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kohei Nakajima
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kenji Shono
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoshifumi Mizobuchi
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Toshitaka Fujihara
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Eiji Shikata
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tadashi Yamaguchi
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keiko Kitazato
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Oltea Sampetrean
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yasushi Takagi
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
27
|
Mellai M, Annovazzi L, Boldorini R, Bertero L, Cassoni P, De Blasio P, Biunno I, Schiffer D. SEL1L plays a major role in human malignant gliomas. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2019; 6:17-29. [PMID: 31111685 PMCID: PMC6966709 DOI: 10.1002/cjp2.134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/30/2019] [Accepted: 05/07/2019] [Indexed: 12/22/2022]
Abstract
Suppressor of Lin-12-like (C. elegans) (SEL1L) participates in the endoplasmic reticulum-associated protein degradation pathway, malignant transformation and stem cell biology. We explored the role of SEL1L in 110 adult gliomas, of different molecular subtype and grade, in relation to cell proliferation, stemness, glioma-associated microglia/macrophages (GAMs), prognostic markers and clinical outcome. SEL1L protein expression was assessed by immunohistochemistry and Western blotting. Genetic and epigenetic alterations were detected by molecular genetics techniques. SEL1L was overexpressed in anaplastic gliomas (World Health Organization [WHO] grade III) and in glioblastoma (GB, WHO grade IV) with the highest labelling index (LI) in the latter. Immunoreactivity was significantly associated with histological grade (p = 0.002) and cell proliferation index Ki-67/MIB-1 (p = 0.0001). In GB, SEL1L co-localised with stemness markers Nestin and Sox2. Endothelial cells and vascular pericytes of proliferative tumour blood vessels expressed SEL1L suggesting a role in tumour neo-vasculature. GAMs consistently expressed SEL1L. SEL1L overexpression was significantly associated with TERT promoter mutations (p = 0.0001), EGFR gene amplification (p = 0.0013), LOH on 10q (p = 0.0012) but was mutually exclusive with IDH1/2 mutations (p = 0.0001). SEL1L immunoreactivity correlated with tumour progression and cell proliferation, conditioning poor patient survival and response to therapy. This study emphasises SEL1L as a potential biomarker for the most common subgroup of TERT mutant/EGFR amplified/IDH-WT GBs.
Collapse
Affiliation(s)
- Marta Mellai
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale "A. Avogadro", Novara, Italy.,Fondazione Edo ed Elvo Tempia Valenta - ONLUS, Biella, Italy
| | - Laura Annovazzi
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Vercelli, Italy
| | - Renzo Boldorini
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Luca Bertero
- Dipartimento di Scienze Mediche, Università degli Studi di Torino/Città della Salute e della Scienza, Torino, Italy
| | - Paola Cassoni
- Dipartimento di Scienze Mediche, Università degli Studi di Torino/Città della Salute e della Scienza, Torino, Italy
| | | | - Ida Biunno
- ISENET Biobanking, Milano, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milano, Italy
| | - Davide Schiffer
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Vercelli, Italy
| |
Collapse
|
28
|
Pro-Inflammatory Activation of A New Immortalized Human Microglia Cell Line. Brain Sci 2019; 9:brainsci9050111. [PMID: 31096716 PMCID: PMC6562668 DOI: 10.3390/brainsci9050111] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 11/25/2022] Open
Abstract
The characterization of human microglia has been hampered by poor availability of human cell sources. However, microglia is involved in the physiopathology of multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, HIV dementia, retinal degenerative diseases, cancer, and many other conditions. Therefore, there is an important need to have experimental paradigms of human microglia characterized and usable to study the role of microglia in the different pathologies in which it is involved. In the present work, we carried out an extensive characterization of Immortalized Human Microglia—SV40 cell line (IMhu), marketed by Applied Biological Material. The functional response of IMhu to a large variety of stimuli was studied. In particular, we investigated morphology, mortality, and changes in the production of different cytokines and chemokines, both under basal conditions and after stimulation. Moreover, western blotting analysis was conducted on phospho-mTOR (Ser 2448) and downstream parameters, p-P70S6K and 4EBP1, in order to understand if IMhu can be used for evaluations of mTOR pathway. In conclusion, IMhu cells proved to be a useful experimental model to investigate the physiopathology of inflammatory disease that involved microglia cells, including pathological conditions that involved the mTOR pathway.
Collapse
|
29
|
Iv M, Samghabadi P, Holdsworth S, Gentles A, Rezaii P, Harsh G, Li G, Thomas R, Moseley M, Daldrup-Link HE, Vogel H, Wintermark M, Cheshier S, Yeom KW. Quantification of Macrophages in High-Grade Gliomas by Using Ferumoxytol-enhanced MRI: A Pilot Study. Radiology 2018; 290:198-206. [PMID: 30398435 DOI: 10.1148/radiol.2018181204] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Purpose To investigate ferumoxytol-enhanced MRI as a noninvasive imaging biomarker of macrophages in adults with high-grade gliomas. Materials and Methods In this prospective study, adults with high-grade gliomas were enrolled between July 2015 and July 2017. Each participant was administered intravenous ferumoxytol (5 mg/kg) and underwent 3.0-T MRI 24 hours later. Two sites in each tumor were selected for intraoperative sampling on the basis of the degree of ferumoxytol-induced signal change. Susceptibility and the relaxation rates R2* (1/T2*) and R2 (1/T2) were obtained by region-of-interest analysis by using the respective postprocessed maps. Each sample was stained with Prussian blue, CD68, CD163, and glial fibrillary acidic protein. Pearson correlation and linear mixed models were performed to assess the relationship between imaging measurements and number of 400× magnification high-power fields with iron-containing macrophages. Results Ten adults (four male participants [mean age, 65 years ± 9 {standard deviation}; age range, 57-74 years] and six female participants [mean age, 53 years ± 12 years; age range, 32-65 years]; mean age of all participants, 58 years ± 12 [age range, 32-74 years]) with high-grade gliomas were included. Significant positive correlations were found between susceptibility, R2*, and R2' and the number of high-power fields with CD163-positive (r range, 0.64-0.71; P < .01) and CD68-positive (r range, 0.55-0.57; P value range, .01-.02) iron-containing macrophages. No significant correlation was found between R2 and CD163-positive (r = 0.33; P = .16) and CD68-positive (r = 0.24; P = .32) iron-containing macrophages. Similar significance results were obtained with linear mixed models. At histopathologic analysis, iron particles were found only in macrophages; none was found in glial fibrillary acidic protein-positive tumor cells. Conclusion MRI measurements of susceptibility, R2*, and R2' (R2* - R2) obtained after ferumoxytol administration correlate with iron-containing macrophage concentration, and this shows their potential as quantitative imaging markers of macrophages in malignant gliomas. © RSNA, 2018 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Michael Iv
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Peyman Samghabadi
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Samantha Holdsworth
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Andrew Gentles
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Paymon Rezaii
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Griffith Harsh
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Gordon Li
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Reena Thomas
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Michael Moseley
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Heike E Daldrup-Link
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Hannes Vogel
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Max Wintermark
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Samuel Cheshier
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| | - Kristen W Yeom
- From the Departments of Radiology (M.I., P.R., H.E.D.L., M.W., K.W.Y.) and Pathology (P.S., H.V.), Stanford University Medical Center, 300 Pasteur Dr, Grant Building, Room S031E, Stanford, CA 94305; Richard M. Lucas Center for Imaging (S.H., M.M.) and Departments of Medicine (Biomedical Informatics Research) (A.G.), Neurosurgery (G.H., G.L., S.C.), and Neurology (Neuro-Oncology) (R.T.), Stanford University, Stanford, Calif
| |
Collapse
|
30
|
Matias D, Balça-Silva J, da Graça GC, Wanjiru CM, Macharia LW, Nascimento CP, Roque NR, Coelho-Aguiar JM, Pereira CM, Dos Santos MF, Pessoa LS, Lima FRS, Schanaider A, Ferrer VP, Moura-Neto V. Microglia/Astrocytes-Glioblastoma Crosstalk: Crucial Molecular Mechanisms and Microenvironmental Factors. Front Cell Neurosci 2018; 12:235. [PMID: 30123112 PMCID: PMC6086063 DOI: 10.3389/fncel.2018.00235] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
In recent years, the functions of glial cells, namely, astrocytes and microglia, have gained prominence in several diseases of the central nervous system, especially in glioblastoma (GB), the most malignant primary brain tumor that leads to poor clinical outcomes. Studies showed that microglial cells or astrocytes play a critical role in promoting GB growth. Based on the recent findings, the complex network of the interaction between microglial/astrocytes cells and GB may constitute a potential therapeutic target to overcome tumor malignancy. In the present review, we summarize the most important mechanisms and functions of the molecular factors involved in the microglia or astrocytes-GB interactions, which is particularly the alterations that occur in the cell's extracellular matrix and the cytoskeleton. We overview the cytokines, chemokines, neurotrophic, morphogenic, metabolic factors, and non-coding RNAs actions crucial to these interactions. We have also discussed the most recent studies regarding the mechanisms of transportation and communication between microglial/astrocytes - GB cells, namely through the ABC transporters or by extracellular vesicles. Lastly, we highlight the therapeutic challenges and improvements regarding the crosstalk between these glial cells and GB.
Collapse
Affiliation(s)
- Diana Matias
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joana Balça-Silva
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences Consortium, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Grazielle C da Graça
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Caroline M Wanjiru
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucy W Macharia
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carla Pires Nascimento
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia R Roque
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Juliana M Coelho-Aguiar
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Marcos F Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana S Pessoa
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Flavia R S Lima
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Schanaider
- Centro de Cirurgia Experimental do Departamento de Cirurgia da Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Valéria P Ferrer
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Universidade do Grande Rio (Unigranrio), Duque de Caxias, Brazil
| |
Collapse
|
31
|
Glioblastoma niches: from the concept to the phenotypical reality. Neurol Sci 2018; 39:1161-1168. [PMID: 29736738 DOI: 10.1007/s10072-018-3408-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/05/2018] [Indexed: 12/24/2022]
Abstract
Recently, the concept of niches as sites of tumor progression, invasion, and angiogenesis in glioblastoma (GB) has been extensively debated. Niches, considered the sites in which glioblastoma stem cells (GSCs) reside, have been classified as perivascular, perinecrotic, and invasive. However, from a neuropathological point of view, it is not easy to establish when a tumor structure can be considered a niche. The relevant literature has been reviewed in the light of our recent experience on the subject. As for perinecrotic niches, the occurrence of GSCs around necrosis is interpreted as triggered by hypoxia through HIF-1α. Our alternative hypothesis is that, together with progenitors, they are the cell constituents of hyper-proliferative areas of GB, where perinecrotic niches have developed, and they would, therefore, represent the remnants of GSCs/progenitors spared by the developing necrosis. Perivascular structures originate from both transport vessels and exchange vessels, i.e., venules, arterioles, or the undefinable neo-formed small vessels, but only those in which a direct contact between GSCs/progenitors and endothelial cells occurs can be called niches. Both pericytes and microglia/macrophages play a role in niche function: Macrophages of blood origin invade GB only after the appearance of "mother vessels" with consequent blood-brain barrier disruption. Not all vessel/tumor cell structures can be considered niches, that is, crucial sites of tumor progression, invasion, and angiogenesis.
Collapse
|