1
|
Licón-Muñoz Y, Avalos V, Subramanian S, Granger B, Martinez F, García-Montaño LA, Varela S, Moore D, Perkins E, Kogan M, Berto S, Chohan MO, Bowers CA, Piccirillo SGM. Single-nucleus and spatial landscape of the sub-ventricular zone in human glioblastoma. Cell Rep 2025; 44:115149. [PMID: 39752252 DOI: 10.1016/j.celrep.2024.115149] [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: 06/05/2024] [Revised: 10/22/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The sub-ventricular zone (SVZ) is the most well-characterized neurogenic area in the mammalian brain. We previously showed that in 65% of patients with glioblastoma (GBM), the SVZ is a reservoir of cancer stem-like cells that contribute to treatment resistance and the emergence of recurrence. Here, we build a single-nucleus RNA-sequencing-based microenvironment landscape of the tumor mass and the SVZ of 15 patients and two histologically normal SVZ samples as controls. We identify a ZEB1-centered mesenchymal signature in the tumor cells of the SVZ. Moreover, the SVZ microenvironment is characterized by tumor-supportive microglia, which spatially coexist and establish crosstalks with tumor cells. Last, differential gene expression analyses, predictions of ligand-receptor and incoming/outgoing interactions, and functional assays reveal that the interleukin (IL)-1β/IL-1RAcP and Wnt-5a/Frizzled-3 pathways represent potential therapeutic targets in the SVZ. Our data provide insights into the biology of the SVZ in patients with GBM and identify potential targets of this microenvironment.
Collapse
Affiliation(s)
- Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Vanessa Avalos
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Suganya Subramanian
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bryan Granger
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Frank Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Leopoldo A García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Samantha Varela
- University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Drew Moore
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Eddie Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Michael Kogan
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM 87131, USA
| | - Stefano Berto
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Muhammad O Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Christian A Bowers
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM 87131, USA
| | - Sara G M Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
2
|
Ding Y, Li SY, Lv W, Li L, Zhang HW, Zhang Z, Zhang YJ, Zhang ZY, Lu XW. Pyroptosis Signature Gene CHMP4B Regulates Microglia Pyroptosis by Inhibiting GSDMD in Alzheimer's Disease. Mol Neurobiol 2025; 62:77-90. [PMID: 38823000 DOI: 10.1007/s12035-024-04255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
In this study, we aimed to work through the key genes involved in the process of pyroptosis in Alzheimer's disease (AD) to identify potential biomarkers using bioinformatics technology and further explore the underlying molecular mechanisms. The transcriptome data of brain tissue in AD patients were screened from the GEO database, and pyroptosis-related genes were analyzed. The functions of differential genes were analyzed by enrichment analysis and protein-protein interaction. The diagnostic model was established using LASSO and logistic regression analysis, and the correlation of clinical data was analyzed. Based on single-cell analysis of brain tissues of patients with AD, immunofluorescence and western blotting were used to explore the key cells affected by the hub gene. After GSEA, qRT-PCR, western blotting, LDH, ROS, and JC-1 were used to investigate the potential mechanism of the hub gene on pyroptosis. A total of 15 pyroptosis differentially expressed genes were identified. A prediction model consisting of six genes was established by LASSO and logistic regression analysis, and the area under the curve was up to 0.81. As a hub gene, CHMP4B was negatively correlated with the severity of AD. CHMP4B expression was decreased in the hippocampal tissue of patients with AD and mice. Single-cell analysis showed that CHMP4B was downregulated in AD microglia. Overexpression of CHMP4B reduced the release of LDH and ROS and restored mitochondrial membrane potential, thereby alleviating the inflammatory response during microglial pyroptosis. In summary, CHMP4B as a hub gene provides a new strategy for the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Yi Ding
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Shi-Yao Li
- Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wei Lv
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lei Li
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Hui-Wen Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhiren Zhang
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Yong-Jie Zhang
- Department of Human Anatomy, Human Brain Bank of Nanjing Medical University, Nanjing, China.
| | - Zhi-Yuan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
- The Key Laboratory of Antibody Technique of the Ministry of Health, Nanjing Medical University, Nanjing, China.
- Department of Neurology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| | - Xiao-Wei Lu
- Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Xiao R, Huang X, Gao S, Duan J, Zhang Y, Zhang M. Microglia in retinal diseases: From pathogenesis towards therapeutic strategies. Biochem Pharmacol 2024; 230:116550. [PMID: 39307318 DOI: 10.1016/j.bcp.2024.116550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/21/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Microglia, a widely dispersed cohort of immune cells in the retina, are intricately involved in a diverse range of pivotal biological processes, including inflammation, vascular development, complement activation, antigen presentation, and phagocytosis. Within the retinal milieu, microglia are crucial for the clearance of dead cells and cellular debris, release of anti-inflammatory agents, and orchestration of vascular network remodeling to maintain homeostasis. In addition, microglia are key mediators of neuroinflammation. Triggered by oxidative stress, elevated intraocular pressure, genetic anomalies, and immune dysregulation, microglia release numerous inflammatory cytokines, contributing to the pathogenesis of various retinal disorders. Recent studies on the ontogeny and broad functions of microglia in the retina have elucidated their characteristics during retinal development, homeostasis, and disease. Furthermore, therapeutic strategies that target microglia and their effector cytokines have been developed and shown positive results for some retinal diseases. Therefore, we systematically review the microglial ontogeny in the retina, elucidate their dual roles in retinal homeostasis and disease pathogenesis, and demonstrate microglia-based targeted therapeutic strategies for retinal diseases.
Collapse
Affiliation(s)
- Ruihan Xiao
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xi Huang
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Sheng Gao
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianan Duan
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yun Zhang
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meixia Zhang
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Zeng L, Zhang HH. Robust brain MRI image classification with SIBOW-SVM. Comput Med Imaging Graph 2024; 118:102451. [PMID: 39515189 DOI: 10.1016/j.compmedimag.2024.102451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Primary Central Nervous System tumors in the brain are among the most aggressive diseases affecting humans. Early detection and classification of brain tumor types, whether benign or malignant, glial or non-glial, is critical for cancer prevention and treatment, ultimately improving human life expectancy. Magnetic Resonance Imaging (MRI) is the most effective technique for brain tumor detection, generating comprehensive brain scans. However, human examination can be error-prone and inefficient due to the complexity, size, and location variability of brain tumors. Recently, automated classification techniques using machine learning methods, such as Convolutional Neural Networks (CNNs), have demonstrated significantly higher accuracy than manual screening. However, deep learning-based image classification methods, including CNNs, face challenges in estimating class probabilities without proper model calibration (Guo et al., 2017; Minderer et al., 2021). In this paper, we propose a novel brain tumor image classification method called SIBOW-SVM, which integrates the Bag-of-Features model with SIFT feature extraction and weighted Support Vector Machines. This new approach can effectively extract hidden image features, enabling differentiation of various tumor types, provide accurate label predictions, and estimate probabilities of images belonging to each class, offering high-confidence classification decisions. We have also developed scalable and parallelable algorithms to facilitate the practical implementation of SIBOW-SVM for massive image datasets. To benchmark our method, we apply SIBOW-SVM to a public dataset of brain tumor MRI images containing four classes: glioma, meningioma, pituitary, and normal. Our results demonstrate that the new method outperforms state-of-the-art techniques, including CNNs, in terms of uncertainty quantification, classification accuracy, computational efficiency, and data robustness.
Collapse
Affiliation(s)
- Liyun Zeng
- Statistics and Data Science GIDP, University of Arizona, Tucson, Arizona 85721, USA
| | - Hao Helen Zhang
- Department of Mathematics, University of Arizona, Tucson, Arizona 85721, USA.
| |
Collapse
|
5
|
Keane L, Cryan JF, Gleeson JP. Exploiting the gut microbiome for brain tumour treatment. Trends Mol Med 2024:S1471-4914(24)00222-3. [PMID: 39256110 DOI: 10.1016/j.molmed.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Increasing evidence suggests that the gut microbiome plays a key role in a host of pathological conditions, including cancer. Indeed, the bidirectional communication that occurs between the gut and the brain, known as the 'gut-brain axis,' has recently been implicated in brain tumour pathology. Here, we focus on current research that supports a gut microbiome-brain tumour link with emphasis on high-grade gliomas, the most aggressive of all brain tumours, and the impact on the glioma tumour microenvironment. We discuss the potential use of gut-brain axis signals to improve responses to current and future therapeutic approaches. We highlight that the success of novel treatment strategies may rely on patient-specific microbiome profiles, and these should be considered for personalised treatment approaches.
Collapse
Affiliation(s)
- Lily Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Jack P Gleeson
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland; CUH/UCC Cancer Centre, Cork University Hospital, Cork, Ireland.
| |
Collapse
|
6
|
Yang Y, Luo X, Wang Y, Xu A, Peng L, Zhang X, Wang Z, Ying Y, Li K. β-Mangostin targets and suppresses glioma via STING activation and tumor-associated microglia polarization. Biomed Pharmacother 2024; 177:117074. [PMID: 38972149 DOI: 10.1016/j.biopha.2024.117074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/09/2024] Open
Abstract
Glioma, a common and highly malignant central nervous system tumor, markedly influences patient prognosis via interactions with glioma-associated macrophages. Previous research has revealed the anticancer potential of β-mangostin, a xanthone derivative obtained from the mangosteen fruit. This research investigated the role of β-mangostin on microglia in the glioma microenvironment and evaluated the efficacy of β-mangostin combined with anti-PD-1 antibody (αPD-1) in glioma-bearing mice. The results showed that, β-mangostin attenuated M2 polarization in BV2 cells and promoted M1-related interleukin (IL)-1β and IL-6 secretion, thereby inhibiting glioma invasion. In addition, β-mangostin improved the anti-glioma effects of αPD-1 and increased CD8+T cell and M1-type microglia infiltration. Mechanistically, β-mangostin bound to the stimulator of interferon genes (STING) protein, which is crucial for the anti-tumor innate immune response, and promoted STING phosphorylation in microglia, both in vivo and in vitro. These results provide insights into its mode of action and supporting further investigation into β-mangostin as a therapeutic agent.
Collapse
Affiliation(s)
- Yimin Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xuling Luo
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yaling Wang
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Aibo Xu
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Lina Peng
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaoting Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang 310063, China.
| | - Youmin Ying
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Kaiqiang Li
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang 310063, China.
| |
Collapse
|
7
|
Rodrigues FDS, Newton WR, Tassinari ID, da Cunha Xavier FH, Marx A, de Fraga LS, Wright K, Guedes RP, Bambini-Jr V. Cannabidiol prevents LPS-induced inflammation by inhibiting the NLRP3 inflammasome and iNOS activity in BV2 microglia cells via CB2 receptors and PPARγ. Neurochem Int 2024; 177:105769. [PMID: 38761855 DOI: 10.1016/j.neuint.2024.105769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Neuroinflammation stands as a critical player in the pathogenesis of diverse neurological disorders, with microglial cells playing a central role in orchestrating the inflammatory landscape within the central nervous system. Cannabidiol (CBD) has gained attention for its potential to elicit anti-inflammatory responses in microglia, offering promising perspectives for conditions associated with neuroinflammation. Here we investigated whether the NLRP3 inflammasome and inducible nitric oxide synthase (iNOS) are involved in the protective effects of CBD, and if their modulation is dependent on cannabinoid receptor 2 (CB2) and PPARγ signalling pathways. We found that treatment with CBD attenuated pro-inflammatory markers in lipopolysaccharide (LPS)-challenged BV2 microglia in a CB2- and PPARγ-dependent manner. At a molecular level, CBD inhibited the LPS-induced pro-inflammatory responses by suppressing iNOS and NLRP3/Caspase-1-dependent signalling cascades, resulting in reduced nitric oxide (NO), interleukin-1β (IL-1β), and tumour necrosis factor-alpha (TNF-α) concentrations. Notably, the protective effects of CBD on NLRP3 expression, Caspase-1 activity, and IL-1β concentration were partially hindered by the antagonism of both CB2 receptors and PPARγ, while iNOS expression and NO secretion were dependent exclusively on PPARγ activation, with no CB2 involvement. Interestingly, CBD exhibited a protective effect against TNF-α increase, regardless of CB2 or PPARγ activation. Altogether, these findings indicate that CB2 receptors and PPARγ mediate the anti-inflammatory effects of CBD on the NLRP3 inflammasome complex, iNOS activity and, ultimately, on microglial phenotype. Our results highlight the specific components responsible for the potential therapeutic applications of CBD on neuroinflammatory conditions.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil; Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| | - William Robert Newton
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom; MRC Centre for Medical Mycology, Exeter University, Exeter, United Kingdom.
| | - Isadora D'Ávila Tassinari
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom; Graduate Program in Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | | | - Adél Marx
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| | - Luciano Stürmer de Fraga
- Graduate Program in Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Karen Wright
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Victorio Bambini-Jr
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, Lancashire, United Kingdom.
| |
Collapse
|
8
|
Zhong J, Xing X, Gao Y, Pei L, Lu C, Sun H, Lai Y, Du K, Xiao F, Yang Y, Wang X, Shi Y, Bai F, Zhang N. Distinct roles of TREM2 in central nervous system cancers and peripheral cancers. Cancer Cell 2024; 42:968-984.e9. [PMID: 38788719 DOI: 10.1016/j.ccell.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/26/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024]
Abstract
Glioblastomas (GBM) are incurable central nervous system (CNS) cancers characterized by substantial myeloid cell infiltration. Whether myeloid cell-directed therapeutic targets identified in peripheral non-CNS cancers are applicable to GBM requires further study. Here, we identify that the critical immunosuppressive target in peripheral cancers, triggering receptor expressed on myeloid cells-2 (TREM2), is immunoprotective in GBM. Genetic or pharmacological TREM2 deficiency promotes GBM progression in vivo. Single-cell and spatial sequencing reveals downregulated TREM2 in GBM-infiltrated myeloid cells. TREM2 negatively correlates with immunosuppressive myeloid and T cell exhaustion signatures in GBM. We further demonstrate that during GBM progression, CNS-enriched sphingolipids bind TREM2 on myeloid cells and elicit antitumor responses. Clinically, high TREM2 expression in myeloid cells correlates with better survival in GBM. Adeno-associated virus-mediated TREM2 overexpression impedes GBM progression and synergizes with anti-PD-1 therapy. Our results reveal distinct functions of TREM2 in CNS cancers and support organ-specific myeloid cell remodeling in cancer immunotherapy.
Collapse
Affiliation(s)
- Jian Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Xudong Xing
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China; Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
| | - Yixin Gao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Lei Pei
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China; Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
| | - Chenfei Lu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Huixin Sun
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Yanxing Lai
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Kang Du
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Feizhe Xiao
- Department of Scientific Research Section, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Ying Yang
- Institute of Pathology and Southwest Cancer Centre, Key Laboratory of Tumor Immunopathology of the Ministry of Education of China, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Yu-Yue Pathology Scientific Research Center and Jinfeng Laboratory, Chongqing 400039, China
| | - Xiuxing Wang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Centre, Key Laboratory of Tumor Immunopathology of the Ministry of Education of China, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Yu-Yue Pathology Scientific Research Center and Jinfeng Laboratory, Chongqing 400039, China
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China; Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China.
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
9
|
Licón-Muñoz Y, Avalos V, Subramanian S, Granger B, Martinez F, Varela S, Moore D, Perkins E, Kogan M, Berto S, Chohan M, Bowers C, Piccirillo S. Single-nucleus and spatial landscape of the sub-ventricular zone in human glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590852. [PMID: 38712234 PMCID: PMC11071523 DOI: 10.1101/2024.04.24.590852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The sub-ventricular zone (SVZ) is the most well-characterized neurogenic area in the mammalian brain. We previously showed that in 65% of patients with glioblastoma (GBM), the SVZ is a reservoir of cancer stem-like cells that contribute to treatment resistance and emergence of recurrence. Here, we built a single-nucleus RNA-sequencing-based microenvironment landscape of the tumor mass (T_Mass) and the SVZ (T_SVZ) of 15 GBM patients and 2 histologically normal SVZ (N_SVZ) samples as controls. We identified a mesenchymal signature in the T_SVZ of GBM patients: tumor cells from the T_SVZ relied on the ZEB1 regulatory network, whereas tumor cells in the T_Mass relied on the TEAD1 regulatory network. Moreover, the T_SVZ microenvironment was predominantly characterized by tumor-supportive microglia, which spatially co-exist and establish heterotypic interactions with tumor cells. Lastly, differential gene expression analyses, predictions of ligand-receptor and incoming/outgoing interactions, and functional assays revealed that the IL-1β/IL-1RAcP and Wnt-5a/Frizzled-3 pathways are therapeutic targets in the T_SVZ microenvironment. Our data provide insights into the biology of the SVZ in GBM patients and identify specific targets of this microenvironment.
Collapse
Affiliation(s)
- Y. Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| | - V. Avalos
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| | - S. Subramanian
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - B. Granger
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - F. Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| | - S. Varela
- University of New Mexico School of Medicine, Albuquerque, NM
| | - D. Moore
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - E. Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS
| | - M. Kogan
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM
| | - S. Berto
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - M.O. Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS
| | - C.A. Bowers
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM
| | - S.G.M. Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| |
Collapse
|
10
|
Khalighi S, Reddy K, Midya A, Pandav KB, Madabhushi A, Abedalthagafi M. Artificial intelligence in neuro-oncology: advances and challenges in brain tumor diagnosis, prognosis, and precision treatment. NPJ Precis Oncol 2024; 8:80. [PMID: 38553633 PMCID: PMC10980741 DOI: 10.1038/s41698-024-00575-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
This review delves into the most recent advancements in applying artificial intelligence (AI) within neuro-oncology, specifically emphasizing work on gliomas, a class of brain tumors that represent a significant global health issue. AI has brought transformative innovations to brain tumor management, utilizing imaging, histopathological, and genomic tools for efficient detection, categorization, outcome prediction, and treatment planning. Assessing its influence across all facets of malignant brain tumor management- diagnosis, prognosis, and therapy- AI models outperform human evaluations in terms of accuracy and specificity. Their ability to discern molecular aspects from imaging may reduce reliance on invasive diagnostics and may accelerate the time to molecular diagnoses. The review covers AI techniques, from classical machine learning to deep learning, highlighting current applications and challenges. Promising directions for future research include multimodal data integration, generative AI, large medical language models, precise tumor delineation and characterization, and addressing racial and gender disparities. Adaptive personalized treatment strategies are also emphasized for optimizing clinical outcomes. Ethical, legal, and social implications are discussed, advocating for transparency and fairness in AI integration for neuro-oncology and providing a holistic understanding of its transformative impact on patient care.
Collapse
Affiliation(s)
- Sirvan Khalighi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kartik Reddy
- Department of Radiology, Emory University, Atlanta, GA, USA
| | - Abhishek Midya
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Krunal Balvantbhai Pandav
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Atlanta Veterans Administration Medical Center, Atlanta, GA, USA.
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA.
- The Cell and Molecular Biology Program, Winship Cancer Institute, Atlanta, GA, USA.
| |
Collapse
|
11
|
Marallano VJ, Ughetta ME, Tejero R, Nanda S, Ramalingam R, Stalbow L, Sattiraju A, Huang Y, Ramakrishnan A, Shen L, Wojcinski A, Kesari S, Zou H, Tsankov AM, Friedel RH. Hypoxia drives shared and distinct transcriptomic changes in two invasive glioma stem cell lines. Sci Rep 2024; 14:7246. [PMID: 38538643 PMCID: PMC10973515 DOI: 10.1038/s41598-024-56102-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/01/2024] [Indexed: 07/12/2024] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant cancer of the central nervous system. Insufficient oxygenation (hypoxia) has been linked to GBM invasion and aggression, leading to poor patient outcomes. Hypoxia induces gene expression for cellular adaptations. However, GBM is characterized by high intertumoral (molecular subtypes) and intratumoral heterogeneity (cell states), and it is not well understood to what extent hypoxia triggers patient-specific gene responses and cellular diversity in GBM. Here, we surveyed eight patient-derived GBM stem cell lines for invasion phenotypes in 3D culture, which identified two GBM lines showing increased invasiveness in response to hypoxia. RNA-seq analysis of the two patient GBM lines revealed a set of shared hypoxia response genes concerning glucose metabolism, angiogenesis, and autophagy, but also a large set of patient-specific hypoxia-induced genes featuring cell migration and anti-inflammation, highlighting intertumoral diversity of hypoxia responses in GBM. We further applied the Shared GBM Hypoxia gene signature to single cell RNA-seq datasets of glioma patients, which showed that hypoxic cells displayed a shift towards mesenchymal-like (MES) and astrocyte-like (AC) states. Interestingly, in response to hypoxia, tumor cells in IDH-mutant gliomas displayed a strong shift to the AC state, whereas tumor cells in IDH-wildtype gliomas mainly shifted to the MES state. This distinct hypoxia response of IDH-mutant gliomas may contribute to its more favorable prognosis. Our transcriptomic studies provide a basis for future approaches to better understand the diversity of hypoxic niches in gliomas.
Collapse
Affiliation(s)
- Valerie J Marallano
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mary E Ughetta
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rut Tejero
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sidhanta Nanda
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rohana Ramalingam
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lauren Stalbow
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anirudh Sattiraju
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yong Huang
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexandre Wojcinski
- Pacific Neuroscience Institute and Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute and Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexander M Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Roland H Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
12
|
Kong Y, Wang D, Jin X, Liu Y, Xu H. Unveiling the significance of TREM1/2 in hemorrhagic stroke: structure, function, and therapeutic implications. Front Neurol 2024; 15:1334786. [PMID: 38385036 PMCID: PMC10879330 DOI: 10.3389/fneur.2024.1334786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
Stroke has long been a major threat to human health worldwide. Hemorrhagic stroke, including intracerebral hemorrhage and subarachnoid hemorrhage, exhibits a high incidence rate and a high mortality and disability rate, imposing a substantial burden on both public health and the economy and society. In recent years, the triggering receptor expressed on myeloid cells (TREM) family has garnered extensive attention in various pathological conditions, including hemorrhagic stroke. This review comprehensively summarizes the structure and function of TREM1/2, as well as their roles and potential mechanisms in hemorrhagic stroke, with the aim of providing guidance for the development of targeted therapeutic strategies in the future.
Collapse
Affiliation(s)
- Yancheng Kong
- Trauma Emergency Center, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Di Wang
- Trauma Emergency Center, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Xu Jin
- Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Yi Liu
- Trauma Emergency Center, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Hui Xu
- Trauma Emergency Center, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| |
Collapse
|
13
|
Wu SY, Yu WJ, Chien TY, Ren YA, Chen CS, Chiang CS. Microglia-mediated drug substance transfer promotes chemoresistance in brain tumors: insights from an in vitro co-culture model using GCV/Tk prodrug system. Cancer Cell Int 2024; 24:35. [PMID: 38238749 PMCID: PMC10795391 DOI: 10.1186/s12935-024-03213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND It is well known that tumor-associated macrophages (TAMs) play essential roles in brain tumor resistance to chemotherapy. However, the detailed mechanisms of how TAMs are involved in brain tumor resistance are still unclear and lack a suitable analysis model. METHODS A BV2 microglial cells with ALTS1C1 astrocytoma cells in vitro co-culture system was used to mimic the microglia dominating tumor stroma in the tumor invasion microenvironment and explore the interaction between microglia and brain tumor cells. RESULTS Our result suggested that microglia could form colonies with glioma cells under high-density culturing conditions and protect glioma cells from apoptosis induced by chemotherapeutic drugs. Moreover, this study demonstrates that microglia could hijack drug substances from the glioma cells and reduce the drug intensity of ALTS1C1 via direct contact. Inhibition of gap junction protein prevented microglial-glioma colony formation and microglia-mediated chemoresistance. CONCLUSIONS This study provides novel insights into how glioma cells acquire chemoresistance via microglia-mediated drug substance transferring, providing a new option for treating chemo-resistant brain tumors.
Collapse
Affiliation(s)
- Sheng-Yan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Wen-Jui Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ting-Yi Chien
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-An Ren
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan.
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, 30013, Taiwan.
- Frontier Research Center On Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
14
|
du Chatinier A, Velilla IQ, Meel MH, Hoving EW, Hulleman E, Metselaar DS. Microglia in pediatric brain tumors: The missing link to successful immunotherapy. Cell Rep Med 2023; 4:101246. [PMID: 37924816 PMCID: PMC10694606 DOI: 10.1016/j.xcrm.2023.101246] [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: 05/02/2023] [Revised: 08/10/2023] [Accepted: 09/26/2023] [Indexed: 11/06/2023]
Abstract
Brain tumors are the leading cause of cancer-related mortality in children. Despite the development of immunotherapeutic strategies for adult brain tumors, progress in pediatric neuro-oncology has been hindered by the complex and poorly understood nature of the brain's immune system during early development, a phase that is critical for the onset of many pediatric brain tumors. A defining characteristic of these tumors is the abundance of microglia, the resident immune cells of the central nervous system. In this review, we explore the concept of microglial diversity across brain regions and throughout development and discuss how their maturation stage may contribute to tumor growth in children. We also summarize the current knowledge on the roles of microglia in common pediatric brain tumor entities and provide examples of myeloid-based immunotherapeutic strategies. Our review underscores the importance of microglial plasticity in pediatric brain tumors and its significance for developing effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Aimée du Chatinier
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Irene Querol Velilla
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Michaël Hananja Meel
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Eelco Wieger Hoving
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Esther Hulleman
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Dennis Serge Metselaar
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands.
| |
Collapse
|
15
|
Cignarella A, Vegeto E, Bolego C, Trabace L, Conti L, Ortona E. Sex-oriented perspectives in immunopharmacology. Pharmacol Res 2023; 197:106956. [PMID: 37820857 DOI: 10.1016/j.phrs.2023.106956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
Several immunopharmacological agents are effective in the treatment of cancer and immune-mediated conditions, with a favorable impact on life expectancy and clinical outcomes for a large number of patients. Nevertheless, response variation and undesirable effects of these drugs represent major issues, and overall efficacy remains unpredictable. Males and females show a distinct difference in immune system responses, with females generally mounting stronger responses to a variety of stimuli. Therefore, exploring sex differences in the efficacy and safety of immunopharmacological agents would strengthen the practice of precision medicine. As a pharmacological target highlight, programmed cell death 1 ligand 1 (PD-L1) is the first functionally characterized ligand of the coinhibitory programmed death receptor 1 (PD-1). The PD-L1/PD-1 crosstalk plays an important role in the immune response and is relevant in cancer, infectious and autoimmune disease. Sex differences in the response to immune checkpoint inhibitors are well documented, with male patients responding better than female patients. Similarly, higher efficacy of and adherence to tumor necrosis factor inhibitors in chronic inflammatory conditions including rheumatoid arthritis and Crohn's disease have been reported in male patients. The pharmacological basis of sex-specific responses to immune system modulating drugs is actively investigated in other settings such as stroke and type 1 diabetes. Advances in therapeutics targeting the endothelium could soon be wielded against autoimmunity and metabolic disorders. Based on the established sexual dimorphism in immune-related pathophysiology and disease presentation, sex-specific immunopharmacological protocols should be integrated into clinical guidelines.
Collapse
Affiliation(s)
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Lucia Conti
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Ortona
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
16
|
Chen X, Deng R, Su D, Ma X, Han X, Wang S, Xia Y, Yang Z, Gong N, Jia Y, Gao X, Ren X. Visual genetic typing of glioma using proximity-anchored in situ spectral coding amplification. EXPLORATION (BEIJING, CHINA) 2023; 3:20220175. [PMID: 37933281 PMCID: PMC10582607 DOI: 10.1002/exp.20220175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/21/2023] [Indexed: 11/08/2023]
Abstract
Gliomas are histologically and genetically heterogeneous tumors. However, classical histopathological typing often ignores the high heterogeneity of tumors and thus cannot meet the requirements of precise pathological diagnosis. Here, proximity-anchored in situ spectral coding amplification (ProxISCA) is proposed for multiplexed imaging of RNA mutations, enabling visual typing of brain gliomas with different pathological grades at the single-cell and tissue levels. The ligation-based padlock probe can discriminate one-nucleotide variations, and the design of proximity primers enables the anchoring of amplicons on target RNA, thus improving localization accuracy. The DNA module-based spectral coding strategy can dramatically improve the multiplexing capacity for imaging RNA mutations through one-time labelling, with low cost and simple operation. One-target-one-amplicon amplification confers ProxISCA the ability to quantify RNA mutation copy number with single-molecule resolution. Based on this approach, it is found that gliomas with higher malignant grades express more genes with high correlation at the cellular and tissue levels and show greater cellular heterogeneity. ProxISCA provides a tool for glioma research and precise diagnosis, which can reveal the relationship between cellular heterogeneity and glioma occurrence or development and assist in pathological prognosis.
Collapse
Affiliation(s)
- Xiaolei Chen
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Ruijie Deng
- College of Biomass Science and EngineeringHealthy Food Evaluation Research CenterSichuan UniversityChengduChina
| | - Dongdong Su
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Xiaochen Ma
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Xu Han
- Institute of High Energy PhysicsChinese Academy of SciencesBeijingChina
| | - Shizheng Wang
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Yuqing Xia
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Zifu Yang
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Ningqiang Gong
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaUSA
| | - Yanwei Jia
- State‐Key Laboratory of Analog and Mixed‐Signal VLSIInstitute of MicroelectronicsUniversity of MacauMacauChina
| | - Xueyun Gao
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Xiaojun Ren
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| |
Collapse
|
17
|
Geraldo LH, Garcia C, Xu Y, Leser FS, Grimaldi I, de Camargo Magalhães ES, Dejaegher J, Solie L, Pereira CM, Correia AH, De Vleeschouwer S, Tavitian B, Canedo NHS, Mathivet T, Thomas JL, Eichmann A, Lima FRS. CCL21-CCR7 signaling promotes microglia/macrophage recruitment and chemotherapy resistance in glioblastoma. Cell Mol Life Sci 2023; 80:179. [PMID: 37314567 DOI: 10.1007/s00018-023-04788-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 06/15/2023]
Abstract
Glioblastoma (GBM) is the most common and fatal primary tumor of the central nervous system (CNS) and current treatments have limited success. Chemokine signaling regulates both malignant cells and stromal cells of the tumor microenvironment (TME), constituting a potential therapeutic target against brain cancers. Here, we investigated the C-C chemokine receptor type 7 (CCR7) and the chemokine (C-C-motif) ligand 21 (CCL21) for their expression and function in human GBM and then assessed their therapeutic potential in preclinical mouse GBM models. In GBM patients, CCR7 expression positively associated with a poor survival. CCL21-CCR7 signaling was shown to regulate tumor cell migration and proliferation while also controlling tumor associated microglia/macrophage recruitment and VEGF-A production, thereby controlling vascular dysmorphia. Inhibition of CCL21-CCR7 signaling led to an increased sensitivity to temozolomide-induced tumor cell death. Collectively, our data indicate that drug targeting of CCL21-CCR7 signaling in tumor and TME cells is a therapeutic option against GBM.
Collapse
Affiliation(s)
- Luiz Henrique Geraldo
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rua César Pernetta, 1.766, Cidade Universitária da UFRJ, Rio de Janeiro, RJ, 21949-590, Brazil.
- Université de Paris, PARCC, INSERM, 75015, Paris, France.
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, 06510-3221, USA.
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510-3221, USA.
| | - Celina Garcia
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rua César Pernetta, 1.766, Cidade Universitária da UFRJ, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Yunling Xu
- Université de Paris, PARCC, INSERM, 75015, Paris, France
| | - Felipe Saceanu Leser
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rua César Pernetta, 1.766, Cidade Universitária da UFRJ, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Izabella Grimaldi
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rua César Pernetta, 1.766, Cidade Universitária da UFRJ, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Eduardo Sabino de Camargo Magalhães
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rua César Pernetta, 1.766, Cidade Universitária da UFRJ, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Joost Dejaegher
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, KU Leuven, Leuven, Belgium
| | - Lien Solie
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, KU Leuven, Leuven, Belgium
| | - Cláudia Maria Pereira
- Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil
| | - Ana Helena Correia
- Departmento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Steven De Vleeschouwer
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, KU Leuven, Leuven, Belgium
| | | | - Nathalie Henriques Silva Canedo
- Departmento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Jean-Leon Thomas
- Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France.
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510-3221, USA.
| | - Anne Eichmann
- Université de Paris, PARCC, INSERM, 75015, Paris, France
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, 06510-3221, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510-3221, USA
| | - Flavia Regina Souza Lima
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rua César Pernetta, 1.766, Cidade Universitária da UFRJ, Rio de Janeiro, RJ, 21949-590, Brazil.
| |
Collapse
|
18
|
Cerneckis J, Shi Y. Modeling brain macrophage biology and neurodegenerative diseases using human iPSC-derived neuroimmune organoids. Front Cell Neurosci 2023; 17:1198715. [PMID: 37342768 PMCID: PMC10277621 DOI: 10.3389/fncel.2023.1198715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, United States
| |
Collapse
|
19
|
Stratoulias V, Ruiz R, Kanatani S, Osman AM, Keane L, Armengol JA, Rodríguez-Moreno A, Murgoci AN, García-Domínguez I, Alonso-Bellido I, González Ibáñez F, Picard K, Vázquez-Cabrera G, Posada-Pérez M, Vernoux N, Tejera D, Grabert K, Cheray M, González-Rodríguez P, Pérez-Villegas EM, Martínez-Gallego I, Lastra-Romero A, Brodin D, Avila-Cariño J, Cao Y, Airavaara M, Uhlén P, Heneka MT, Tremblay MÈ, Blomgren K, Venero JL, Joseph B. ARG1-expressing microglia show a distinct molecular signature and modulate postnatal development and function of the mouse brain. Nat Neurosci 2023:10.1038/s41593-023-01326-3. [PMID: 37169859 DOI: 10.1038/s41593-023-01326-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
Molecular diversity of microglia, the resident immune cells in the CNS, is reported. Whether microglial subsets characterized by the expression of specific proteins constitute subtypes with distinct functions has not been fully elucidated. Here we describe a microglial subtype expressing the enzyme arginase-1 (ARG1; that is, ARG1+ microglia) that is found predominantly in the basal forebrain and ventral striatum during early postnatal mouse development. ARG1+ microglia are enriched in phagocytic inclusions and exhibit a distinct molecular signature, including upregulation of genes such as Apoe, Clec7a, Igf1, Lgals3 and Mgl2, compared to ARG1- microglia. Microglial-specific knockdown of Arg1 results in deficient cholinergic innervation and impaired dendritic spine maturation in the hippocampus where cholinergic neurons project, which in turn results in impaired long-term potentiation and cognitive behavioral deficiencies in female mice. Our results expand on microglia diversity and provide insights into microglia subtype-specific functions.
Collapse
Affiliation(s)
- Vassilis Stratoulias
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Rocío Ruiz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Seville, Spain
| | - Shigeaki Kanatani
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ahmed M Osman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Lily Keane
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Jose A Armengol
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Antonio Rodríguez-Moreno
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Adriana-Natalia Murgoci
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Irene García-Domínguez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Seville, Spain
| | - Isabel Alonso-Bellido
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Seville, Spain
| | - Fernando González Ibáñez
- Department of Molecular Medicine, Université Laval, and Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Laval, Quebec, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Katherine Picard
- Department of Molecular Medicine, Université Laval, and Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Laval, Quebec, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Guillermo Vázquez-Cabrera
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Seville, Spain
| | - Mercedes Posada-Pérez
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Seville, Spain
| | - Nathalie Vernoux
- Department of Molecular Medicine, Université Laval, and Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Laval, Quebec, Canada
| | - Dario Tejera
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Kathleen Grabert
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | | | - Eva M Pérez-Villegas
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Irene Martínez-Gallego
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | | | - David Brodin
- Bioinformatics and Expression Analysis Core Facility, Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Javier Avila-Cariño
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, Sweden
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mikko Airavaara
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marie-Ève Tremblay
- Department of Molecular Medicine, Université Laval, and Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Laval, Quebec, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Jose L Venero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Seville, Spain
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
20
|
Duan J, Wang Y. Modeling nervous system tumors with human stem cells and organoids. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:4. [PMID: 36854987 PMCID: PMC9975125 DOI: 10.1186/s13619-022-00150-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 11/05/2022] [Indexed: 03/02/2023]
Abstract
Nervous system cancers are the 10th leading cause of death worldwide, many of which are difficult to diagnose and exhibit varying degrees of treatment resistance. The limitations of existing cancer models, such as patient-derived xenograft (PDX) models and genetically engineered mouse (GEM) models, call for the development of novel preclinical cancer models to more faithfully mimic the patient's cancer and offer additional insights. Recent advances in human stem cell biology, organoid, and genome-editing techniques allow us to model nervous system tumors in three types of next-generation tumor models: cell-of-origin models, tumor organoids, and 3D multicellular coculture models. In this review, we introduced and compared different human stem cell/organoid-derived models, and comprehensively summarized and discussed the recently developed models for various primary tumors in the central and peripheral nervous systems, including glioblastoma (GBM), H3K27M-mutant Diffuse Midline Glioma (DMG) and H3G34R-mutant High-grade Glioma (HGG), Low-grade Glioma (LGG), Neurofibromatosis Type 1 (NF1), Neurofibromatosis Type 2 (NF2), Medulloblastoma (MB), Atypical Teratoid/rhabdoid Tumor (AT/RT), and meningioma. We further compared these models with PDX and GEM models, and discussed the opportunities and challenges of precision nervous cancer modeling with human stem cells and organoids.
Collapse
Affiliation(s)
- Jie Duan
- grid.412901.f0000 0004 1770 1022Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041 China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
21
|
Dean PT, Hooks SB. Pleiotropic effects of the COX-2/PGE2 axis in the glioblastoma tumor microenvironment. Front Oncol 2023; 12:1116014. [PMID: 36776369 PMCID: PMC9909545 DOI: 10.3389/fonc.2022.1116014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 12/19/2022] [Indexed: 01/27/2023] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive form of malignant glioma. The GBM tumor microenvironment (TME) is a complex ecosystem of heterogeneous cells and signaling factors. Glioma associated macrophages and microglia (GAMs) constitute a significant portion of the TME, suggesting that their functional attributes play a crucial role in cancer homeostasis. In GBM, an elevated GAM population is associated with poor prognosis and therapeutic resistance. Neoplastic cells recruit these myeloid populations through release of chemoattractant factors and dysregulate their induction of inflammatory programs. GAMs become protumoral advocates through production a variety of cytokines, inflammatory mediators, and growth factors that can drive cancer proliferation, invasion, immune evasion, and angiogenesis. Among these inflammatory factors, cyclooxygenase-2 (COX-2) and its downstream product, prostaglandin E2 (PGE2), are highly enriched in GBM and their overexpression is positively correlated with poor prognosis in patients. Both tumor cells and GAMs have the ability to signal through the COX-2 PGE2 axis and respond in an autocrine/paracrine manner. In the GBM TME, enhanced signaling through the COX-2/PGE2 axis leads to pleotropic effects that impact GAM dynamics and drive tumor progression.
Collapse
|
22
|
Microglia and Brain Macrophages as Drivers of Glioma Progression. Int J Mol Sci 2022; 23:ijms232415612. [PMID: 36555253 PMCID: PMC9779147 DOI: 10.3390/ijms232415612] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Evidence is accumulating that the tumour microenvironment (TME) has a key role in the progression of gliomas. Non-neoplastic cells in addition to the tumour cells are therefore finding increasing attention. Microglia and other glioma-associated macrophages are at the centre of this interest especially in the context of therapeutic considerations. New ideas have emerged regarding the role of microglia and, more recently, blood-derived brain macrophages in glioblastoma (GBM) progression. We are now beginning to understand the mechanisms that allow malignant glioma cells to weaken microglia and brain macrophage defence mechanisms. Surface molecules and cytokines have a prominent role in microglia/macrophage-glioma cell interactions, and we discuss them in detail. The involvement of exosomes and microRNAs forms another focus of this review. In addition, certain microglia and glioma cell pathways deserve special attention. These "synergistic" (we suggest calling them "Janus") pathways are active in both glioma cells and microglia/macrophages where they act in concert supporting malignant glioma progression. Examples include CCN4 (WISP1)/Integrin α6β1/Akt and CHI3L1/PI3K/Akt/mTOR. They represent attractive therapeutic targets.
Collapse
|
23
|
Peng L, Hu G, Yao Q, Wu J, He Z, Law BYK, Hu G, Zhou X, Du J, Wu A, Yu L. Microglia autophagy in ischemic stroke: A double-edged sword. Front Immunol 2022; 13:1013311. [PMID: 36466850 PMCID: PMC9708732 DOI: 10.3389/fimmu.2022.1013311] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/25/2022] [Indexed: 08/14/2023] Open
Abstract
Ischemic stroke (IS) is one of the major types of cerebrovascular diseases causing neurological morbidity and mortality worldwide. In the pathophysiological process of IS, microglia play a beneficial role in tissue repair. However, it could also cause cellular damage, consequently leading to cell death. Inflammation is characterized by the activation of microglia, and increasing evidence showed that autophagy interacts with inflammation through regulating correlative mediators and signaling pathways. In this paper, we summarized the beneficial and harmful effects of microglia in IS. In addition, we discussed the interplay between microglia autophagy and ischemic inflammation, as along with its application in the treatment of IS. We believe this could help to provide the theoretical references for further study into IS and treatments in the future.
Collapse
Affiliation(s)
- Li Peng
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
- Department of Medicine Imaging, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Guangqiang Hu
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Qianfang Yao
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ziyang He
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Guishan Hu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaogang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Junrong Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
- Department of Medicine Imaging, School of Clinical Medicine, Southwest Medical University, Luzhou, China
- Department of Chemistry, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Maier AD. Malignant meningioma. APMIS 2022; 130 Suppl 145:1-58. [DOI: 10.1111/apm.13276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Andrea Daniela Maier
- Department of Neurosurgery, Rigshospitalet Copenhagen University Hospital Copenhagen Denmark
- Department of Pathology, Rigshospitalet Copenhagen University Hospital Copenhagen Denmark
| |
Collapse
|
25
|
Maier AD, Meddis A, Mirian C, Haslund-Vinding J, Bartek J, Krog SM, Nguyen TUP, Areškevičiūtė A, Melchior LC, Heegaard S, Kristensen BW, Munch TN, Fugleholm K, Ziebell M, Raleigh DR, Poulsen FR, Gerds TA, Litman T, Scheie D, Mathiesen T. Gene expression analysis during progression of malignant meningioma compared to benign meningioma. J Neurosurg 2022; 138:1302-1312. [PMID: 36115056 DOI: 10.3171/2022.7.jns22585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/22/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Meningioma is the most common primary intracranial neoplasm. Only 1%-3% of meningiomas are malignant according to the 2016 WHO criteria (WHO grade III). High-grade meningiomas present specific gene expression signatures indicating aggressive growth or recurrence. However, changes in gene expression and in neuroinflammatory gene expression signatures in WHO grade III meningiomas and during progression from WHO grade I or II to grade III are unknown. METHODS The authors used a NanoString targeted gene expression panel with focus on 787 genes relevant in meningioma pathology and neuroinflammatory pathways to investigate patients with grade III meningiomas treated at Rigshospitalet from 2000 to 2020 (n = 51). A temporal dimension was added to the investigation by including samples from patients' earlier grade I and II meningiomas and grade III recurrences (n = 139 meningiomas). The authors investigated changes in neuroinflammatory gene expression signatures in 1) grade I meningiomas that later transformed into grade III meningiomas, and 2) grade III meningiomas compared with nonrecurrent grade I meningiomas. RESULTS The authors' data indicate that FOXM1, TOP2A, BIRC5, and MYBL2 were enriched and the HOTAIR regulatory pathway was enriched in grade III meningiomas compared with nonrecurrent grade I meningiomas. They discovered a separation of malignant and benign meningiomas based only on genes involved in microglia regulation with enrichment of P2RY12 in grade I compared with grade III meningiomas. Interestingly, FOXM1 was upregulated in premalignant grade I meningioma years before the grade III transformation. CONCLUSIONS The authors found gene expression changes in low-grade meningiomas that predated histological transformation to grade III meningiomas. Neuroinflammation genes distinguished grade III from grade I meningiomas.
Collapse
Affiliation(s)
- Andrea D Maier
- Departments of1Neurosurgery and.,2Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Alessandra Meddis
- 3Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Jiri Bartek
- Departments of1Neurosurgery and.,4Department of Neurosurgery, Karolinska University Hospital, Solna, Stockholm, Sweden.,5Department of Clinical Neuroscience, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Sebastian M Krog
- 6Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | | | - Aušrinė Areškevičiūtė
- 7Department of Pathology, Danish Reference Center for Prion Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Linea C Melchior
- 2Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Steffen Heegaard
- 2Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,8Department of Ophthalmology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Bjarne W Kristensen
- 9Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.,10Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tina N Munch
- Departments of1Neurosurgery and.,11Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.,17Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | - David R Raleigh
- Departments of12Neurological Surgery and.,13Radiation Oncology, University of California, San Francisco, California
| | - Frantz R Poulsen
- 14Department of Neurosurgery, Odense University Hospital, Odense, Denmark.,15Clinical Institute and BRIDGE, University of Southern Denmark, Odense, Denmark; and
| | - Thomas A Gerds
- 3Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | | | - David Scheie
- 2Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tiit Mathiesen
- Departments of1Neurosurgery and.,17Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Pan Y, Monje M. Neuron-Glial Interactions in Health and Brain Cancer. Adv Biol (Weinh) 2022; 6:e2200122. [PMID: 35957525 PMCID: PMC9845196 DOI: 10.1002/adbi.202200122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/21/2022] [Indexed: 01/28/2023]
Abstract
Brain tumors are devastating diseases of the central nervous system. Brain tumor pathogenesis depends on both tumor-intrinsic oncogenic programs and extrinsic microenvironmental factors, including neurons and glial cells. Glial cells (oligodendrocytes, astrocytes, and microglia) make up half of the cells in the brain, and interact with neurons to modulate neurodevelopment and plasticity. Many brain tumor cells exhibit transcriptomic profiles similar to macroglial cells (oligodendrocytes and astrocytes) and their progenitors, making them likely to subvert existing neuron-glial interactions to support tumor pathogenesis. For example, oligodendrocyte precursor cells, a putative glioma cell of origin, can form bona fide synapses with neurons. Such synapses are recently identified in gliomas and drive glioma pathophysiology, underscoring how brain tumor cells can take advantage of neuron-glial interactions to support cancer progression. In this review, it is briefly summarized how neurons and their activity normally interact with glial cells and glial progenitors, and it is discussed how brain tumor cells utilize neuron-glial interactions to support tumor initiation and progression. Unresolved questions on these topics and potential avenues to therapeutically target neuron-glia-cancer interactions in the brain are also pointed out.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center,co-corresponding: ;
| | - Michelle Monje
- Department of Neurology, Stanford University,Howard Hughes Medical Institute, Stanford University,co-corresponding: ;
| |
Collapse
|
27
|
Li M, Xu B, Li X, Li Y, Qiu S, Chen K, Liu Z, Ding Y, Wang H, Xu J, Wang H. Mitofusin 2 confers the suppression of microglial activation by cannabidiol: Insights from in vitro and in vivo models. Brain Behav Immun 2022; 104:155-170. [PMID: 35688339 DOI: 10.1016/j.bbi.2022.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/12/2022] [Accepted: 06/05/2022] [Indexed: 10/18/2022] Open
Abstract
Currently, there is increasing attention on the regulatory effects of cannabidiol (CBD) on the inflammatory response and the immune system. However, the mechanisms have not yet been completely revealed. Mitofusin 2 (Mfn2) is a mitochondrial fusion protein involved in the inflammatory response. Here, we investigated whether Mfn2 confers the anti-inflammatory effects of CBD. We found that treatment with CBD decreased the levels of tumor necrosis factor α, interleukin 6, inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and ionized calcium-binding adaptor molecule-1 (Iba1) in lipopolysaccharide (LPS)-challenged microglia. CBD also significantly suppressed the increase in reactive oxygen species (ROS) and the decline of mitochondrial membrane potential in BV-2 cells subjected to LPS. Interestingly, CBD treatment increased the expression of Mfn2, while knockdown of Mfn2 blocked the effect of CBD. By contrast, overexpression of Mfn2 reversed the increase in the levels of iNOS, COX-2, and Iba1 induced by Mfn2 small interfering RNA. In mice challenged with LPS, we found that CBD ameliorated the anxiety responses and cognitive deficits, increased the level of Mfn2, and decreased the expression of Iba1. Since neuro-inflammation and microglial activation are the common events that are observed in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, we treated EAE mice with CBD. Mice that received CBD showed amelioration of clinical signs, reduced inflammatory response, and increased myelin basic protein level. Most importantly, the adeno-associated virus delivery of short hairpin RNA against Mfn2 reversed the protective effects of CBD. Altogether, these results indicate that Mfn2 is an essential immunomodulator conferring the anti-inflammatory effects of CBD. Our results also shed new light on the mechanisms underlying the protective effects of CBD against inflammatory diseases including multiple sclerosis.
Collapse
Affiliation(s)
- Mengfan Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bingtian Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xing Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yueqi Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuqin Qiu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Kechun Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhuhe Liu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuewen Ding
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Honghao Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiangping Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao, Greater Bay Area, China.
| | - Haitao Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao, Greater Bay Area, China.
| |
Collapse
|
28
|
Directed evolution of adeno-associated virus for efficient gene delivery to microglia. Nat Methods 2022; 19:976-985. [PMID: 35879607 DOI: 10.1038/s41592-022-01547-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 06/10/2022] [Indexed: 12/12/2022]
Abstract
As the resident immune cells in the central nervous system (CNS), microglia orchestrate immune responses and dynamically sculpt neural circuits in the CNS. Microglial dysfunction and mutations of microglia-specific genes have been implicated in many diseases of the CNS. Developing effective and safe vehicles for transgene delivery into microglia will facilitate the studies of microglia biology and microglia-associated disease mechanisms. Here, we report the discovery of adeno-associated virus (AAV) variants that mediate efficient in vitro and in vivo microglial transduction via directed evolution of the AAV capsid protein. These AAV-cMG and AAV-MG variants are capable of delivering various genetic payloads into microglia with high efficiency, and enable sufficient transgene expression to support fluorescent labeling, Ca2+ and neurotransmitter imaging and genome editing in microglia in vivo. Furthermore, single-cell RNA sequencing shows that the AAV-MG variants mediate in vivo transgene delivery without inducing microglia immune activation. These AAV variants should facilitate the use of various genetically encoded sensors and effectors in the study of microglia-related biology.
Collapse
|
29
|
Yin W, Ping YF, Li F, Lv SQ, Zhang XN, Li XG, Guo Y, Liu Q, Li TR, Yang LQ, Yang KD, Liu YQ, Luo CH, Luo T, Wang WY, Mao M, Luo M, He ZC, Cao MF, Chen C, Miao JY, Zeng H, Wang C, Zhou L, Yang Y, Yang X, Wang QH, Feng H, Shi Y, Bian XW. A map of the spatial distribution and tumour-associated macrophage states in glioblastoma and grade-4 IDH-mutant astrocytoma. J Pathol 2022; 258:121-135. [PMID: 35723032 DOI: 10.1002/path.5984] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/08/2022] [Accepted: 06/16/2022] [Indexed: 11/10/2022]
Abstract
Tumour-associated macrophages (TAMs) abundantly infiltrate high-grade gliomas and orchestrate immune response, but their diversity in isocitrate dehydrogenase (IDH)-differential grade-4 gliomas remains largely unknown. This study aimed to dissect the transcriptional states, spatial distribution and clinicopathological significance of distinct monocyte-derived TAM (Mo-TAM) and microglia-derived TAM (Mg-TAM) clusters across glioblastoma-IDH-wildtype and astrocytoma-IDH-mutant-grade 4 (Astro-IDH-mut-G4). Single-cell RNA sequencing was performed on four cases of human glioblastoma and three cases of Astro-IDH-mut-G4. Cell clustering, single-cell regulatory network inference and gene set enrichment analysis were performed to characterize the functional states of myeloid clusters. Spatial distribution of TAM subsets was determined in human glioma tissues using multiplex immunostaining. The prognostic value of different TAM-cluster specific geneset was evaluated in the TCGA glioma cohort. Profiling and unbiased clustering of 24,227 myeloid cells from glioblastoma and Astro-IDH-mut-G4 identified 9 myeloid cell clusters including monocyte, six Mo/Mg-TAM subsets, dendritic cell, and proliferative myeloid cluster. Different Mo/Mg-TAM clusters manifest functional and transcriptional diversity controlled by specific regulons. Multiplex immunostaining of subset-specific markers identified spatial enrichment of distinct TAM clusters at peri-vascular/necrotic areas in tumour parenchyma or at tumour-brain interface. Glioblastoma harboured a substantially higher number of monocytes and Mo-TAM-inflammatory cluster, whereas Astro-IDH-mut-G4 was with higher proportion of TAM subset mediating antigen presentation. Glioblastomas with higher proportion of monocytes exhibited a mesenchymal signature, increased angiogenesis and worse patient outcome. Our findings provide insight into myeloid cell diversity and its clinical relevance in IDH-differential grade-4 gliomas, and may serve as a resource for immunotherapy development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Wen Yin
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Fei Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xiao-Ning Zhang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Xue-Gang Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Ying Guo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Tian-Ran Li
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Liu-Qing Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Kai-Di Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Yu-Qi Liu
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Chun-Hua Luo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Tao Luo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Wen-Ying Wang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Min Mao
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Min Luo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Mian-Fu Cao
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Cong Chen
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Jing-Ya Miao
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Hui Zeng
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Chao Wang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Lei Zhou
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Ying Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Xi Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Qiang-Hu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| |
Collapse
|
30
|
Zi H, Tuo Z, He Q, Meng J, Hu Y, Li Y, Yang K. Comprehensive Bioinformatics Analysis of Gasdermin Family of Glioma. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:9046507. [PMID: 35463276 PMCID: PMC9033320 DOI: 10.1155/2022/9046507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/07/2022] [Accepted: 03/12/2022] [Indexed: 12/20/2022]
Abstract
Pyroptosis is a programmed cell death mediated by gasdermins (GSDMs). The prognostic value of pyroptosis-related genes in different tumor types has been gradually demonstrated recently. However, the prognostic impact of GSDMs expression in glioma remains unclear. Here, we present a comprehensive bioinformatic analysis of gasdermin family member gene expression, producing a prognostic model for glioma and creating a competing endogenous RNA (ceRNA) network. The mRNA expression profiles and clinical information of glioma patients were downloaded from TCGA and CGGA. A risk score based on the gasdermin family was constructed in the TCGA cohort and validated in CGGA. The Jurkat cell was used to verify the relationship between pyroptosis and activation-induced cell death (AICD). We identify a significant association between the expression of GSDMD and GSDME and the glioma stage. The least absolute shrinkage and selection operator (LASSO) Cox regression analysis was used to construct a prognostic gene model based on the four prognostic gasdermin family genes (GSDMC, GSDMD, GSDME, and PJVK). This model was able to predict the overall survival of glioma patients with high accuracy. We show that gasdermin family genes are expressed primarily by immune cells, endothelial cells, and neuronal cells in the tumor microenvironment, rather than by malignant tumor cells. T cells were significantly activated in high-risk patients; however, the activation-induced cell death (AICD) pathway was also significantly activated, suggesting widespread expiration of cytotoxic T lymphocytes (CTLs), facilitating tumor progression. We also identify the lncRNA/miR-296-5p/GSDMD regulatory axis as an important player in glioma progression. We have conducted a comprehensive bioinformatic analysis identifying the importance of gasdermin family members in glioma; a prognostic algorithm containing four genes was constructed.
Collapse
Affiliation(s)
- Huaduan Zi
- Cancer Center Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhan Tuo
- Cancer Center Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianyuan He
- Cancer Center Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingshu Meng
- Cancer Center Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Hu
- Cancer Center Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Li
- Cancer Center Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kunyu Yang
- Cancer Center Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
31
|
Salvalaggio A, Silvestri E, Sansone G, Pinton L, Magri S, Briani C, Anglani M, Lombardi G, Zagonel V, Della Puppa A, Mandruzzato S, Corbetta M, Bertoldo A. Magnetic Resonance Imaging Correlates of Immune Microenvironment in Glioblastoma. Front Oncol 2022; 12:823812. [PMID: 35392230 PMCID: PMC8980808 DOI: 10.3389/fonc.2022.823812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background Glioblastoma (GBM) is the most commonly occurring primary malignant brain tumor, and it carries a dismal prognosis. Focusing on the tumor microenvironment may provide new insights into pathogenesis, but no clinical tools are available to do this. We hypothesized that the infiltration of different leukocyte populations in the tumoral and peritumoral brain tissues may be measured by magnetic resonance imaging (MRI). Methods Pre-operative MRI was combined with immune phenotyping of intraoperative tumor tissue based on flow cytometry of myeloid cell populations that are associated with immune suppression, namely, microglia and bone marrow-derived macrophages (BMDM). These cell populations were measured from the central and marginal areas of the lesion identified intraoperatively with 5-aminolevulinic acid-guided surgery. MRI features (volume, mean and standard deviation of signal intensity, and fractality) were derived from all MR sequences (T1w, Gd+ T1w, T2w, FLAIR) and ADC MR maps and from different tumor areas (contrast- and non-contrast-enhancing tumor, necrosis, and edema). The principal components of MRI features were correlated with different myeloid cell populations by Pearson's correlation. Results We analyzed 126 samples from 62 GBM patients. The ratio between BMDM and microglia decreases significantly from the central core to the periphery. Several MRI-derived principal components were significantly correlated (p <0.05, r range: [-0.29, -0.41]) with the BMDM/microglia ratio collected in the central part of the tumor. Conclusions We report a significant correlation between structural MRI clinical imaging and the ratio of recruited vs. resident macrophages with different immunomodulatory activities. MRI features may represent a novel tool for investigating the microenvironment of GBM.
Collapse
Affiliation(s)
- Alessandro Salvalaggio
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Erica Silvestri
- Padova Neuroscience Center, University of Padova, Padova, Italy.,Department of Information Engineering, University of Padova, Padova, Italy
| | - Giulio Sansone
- Department of Neuroscience, University of Padova, Padova, Italy
| | - Laura Pinton
- Veneto Institute of Oncology - Istituto di Ricovero e Cura a Carattere Scientifico (IOV-IRCCS), Padova, Italy
| | - Sara Magri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Chiara Briani
- Department of Neuroscience, University of Padova, Padova, Italy
| | | | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Alessandro Della Puppa
- Neurosurgery, Department of NEUROFARBA, University Hospital of Careggi, University of Florence, Florence, Italy
| | - Susanna Mandruzzato
- Veneto Institute of Oncology - Istituto di Ricovero e Cura a Carattere Scientifico (IOV-IRCCS), Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Maurizio Corbetta
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Fondazione Biomedica, Padova, Italy
| | - Alessandra Bertoldo
- Padova Neuroscience Center, University of Padova, Padova, Italy.,Department of Information Engineering, University of Padova, Padova, Italy
| |
Collapse
|
32
|
Single-Cell Molecular Characterization to Partition the Human Glioblastoma Tumor Microenvironment Genetic Background. Cells 2022; 11:cells11071127. [PMID: 35406690 PMCID: PMC8998055 DOI: 10.3390/cells11071127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Glioblastoma (GB) is a devastating primary brain malignancy. The recurrence of GB is inevitable despite the standard treatment of surgery, chemotherapy, and radiation, and the median survival is limited to around 15 months. The barriers to treatment include the complex interactions among the different cellular components inhabiting the tumor microenvironment. The complex heterogeneous nature of GB cells is helped by the local inflammatory tumor microenvironment, which mostly induces tumor aggressiveness and drug resistance. Methods: By using fluorescent multiple labeling and a DEPArray cell separator, we recovered several single cells or groups of single cells from populations of different origins from IDH-WT GB samples. From each GB sample, we collected astrocytes-like (GFAP+), microglia-like (IBA1+), stem-like cells (CD133+), and endothelial-like cells (CD105+) and performed Copy Number Aberration (CNA) analysis with a low sequencing depth. The same tumors were subjected to a bulk CNA analysis. Results: The tumor partition in its single components allowed single-cell molecular subtyping which revealed new aspects of the GB altered genetic background. Conclusions: Nowadays, single-cell approaches are leading to a new understanding of GB physiology and disease. Moreover, single-cell CNAs resource will permit new insights into genome heterogeneity, mutational processes, and clonal evolution in malignant tissues.
Collapse
|
33
|
Zhou K, Harris RA, Shen X. Editorial: Microglia as a Therapeutic Target for Brain Repair: Opportunities and Challenges. Front Cell Neurosci 2022; 16:877567. [PMID: 35370558 PMCID: PMC8965838 DOI: 10.3389/fncel.2022.877567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Kai Zhou
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- *Correspondence: Kai Zhou
| | - Robert Adam Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Xianli Shen
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Immunology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
34
|
|
35
|
Borch JDS, Haslund-Vinding J, Vilhardt F, Maier AD, Mathiesen T. Meningioma-Brain Crosstalk: A Scoping Review. Cancers (Basel) 2021; 13:4267. [PMID: 34503077 PMCID: PMC8428351 DOI: 10.3390/cancers13174267] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background: In recent years, it has become evident that the tumoral microenvironment (TME) plays a key role in the pathogenesis of various cancers. In meningiomas, however, the TME is poorly understood, and it is unknown if glia cells contribute to meningioma growth and behaviour. Objective: This scoping review investigates if the literature describes and substantiates tumour-brain crosstalk in meningiomas and summarises the current evidence regarding the role of the brain parenchyma in the pathogenesis of meningiomas. Methods: We identified studies through the electronic database PubMed. Articles describing glia cells and cytokines/chemokines in meningiomas were selected and reviewed. Results: Monocytes were detected as the most abundant infiltrating immune cells in meningiomas. Only brain-invasive meningiomas elicited a monocytic response at the tumour-brain interface. The expression of cytokines/chemokines in meningiomas has been studied to some extent, and some of them form autocrine loops in the tumour cells. Paracrine interactions between tumour cells and glia cells have not been explored. Conclusion: It is unknown to what extent meningiomas elicit an immune response in the brain parenchyma. We speculate that tumour-brain crosstalk might only be relevant in cases of invasive meningiomas that disrupt the pial-glial basement membrane.
Collapse
Affiliation(s)
- Josefine de Stricker Borch
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (J.d.S.B.); (A.D.M.); (T.M.)
| | - Jeppe Haslund-Vinding
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (J.d.S.B.); (A.D.M.); (T.M.)
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Andrea Daniela Maier
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (J.d.S.B.); (A.D.M.); (T.M.)
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Tiit Mathiesen
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (J.d.S.B.); (A.D.M.); (T.M.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|