1
|
Chi J, Gao Q, Liu D. Tissue-Resident Macrophages in Cancer: Friend or Foe? Cancer Med 2024; 13:e70387. [PMID: 39494816 PMCID: PMC11533131 DOI: 10.1002/cam4.70387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Macrophages are essential in maintaining homeostasis, combating infections, and influencing the process of various diseases, including cancer. Macrophages originate from diverse lineages: Notably, tissue-resident macrophages (TRMs) differ from hematopoietic stem cells and circulating monocyte-derived macrophages based on genetics, development, and function. Therefore, understanding the recruited and TRM populations is crucial for investigating disease processes. METHODS By searching literature databses, we summarized recent relevant studies. Research has shown that tumor-associated macrophages (TAMs) of distinct origins accumulate in tumor microenvironment (TME), with TRM-derived TAMs closely resembling gene signatures of normal TRMs. RESULTS Recent studies have revealed that TRMs play a crucial role in cancer progression. However, organ-specific effects complicate TRM investigations. Nonetheless, the precise involvement of TRMs in tumors is unclear. This review explores the multifaceted roles of TRMs in cancer, presenting insights into their origins, proliferation, the latest research methodologies, their impact across various tumor sites, their potential and strategies as therapeutic targets, interactions with other cells within the TME, and the internal heterogeneity of TRMs. CONCLUSIONS We believe that a comprehensive understanding of the multifaceted roles of TRMs will pave the way for targeted TRM therapies in the treatment of cancer.
Collapse
Affiliation(s)
- Jianhua Chi
- Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and MetastasisTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Qinglei Gao
- Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and MetastasisTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Dan Liu
- Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and MetastasisTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
2
|
Chen Z, Ford KP, Islam MBAR, Wan H, Han H, Ramakrishnan A, Brown RJ, Villanueva V, Wang Y, Davis BT, Weiss C, Cui W, Gate D, Schwulst SJ. Anti-CD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8 + T-cell cytotoxicity after traumatic brain injury. J Neuroinflammation 2024; 21:267. [PMID: 39427160 PMCID: PMC11491007 DOI: 10.1186/s12974-024-03257-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people.
Collapse
Affiliation(s)
- Zhangying Chen
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Kacie P Ford
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mecca B A R Islam
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hanxiao Wan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hyebin Han
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Abhirami Ramakrishnan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ryan J Brown
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Veronica Villanueva
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yidan Wang
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Booker T Davis
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Craig Weiss
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Weiguo Cui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Steven J Schwulst
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
3
|
Duan J, Lv A, Guo Z, Liu Q, Tian C, Yang Y, Bi J, Yu X, Peng G, Luo B, Cai Z, Xu B, Fu Y, Zhang J. CX3CR1 +/UCHL1 + microglial extracellular vesicles in blood: a potential biomarker for multiple sclerosis. J Neuroinflammation 2024; 21:254. [PMID: 39385200 PMCID: PMC11465848 DOI: 10.1186/s12974-024-03243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
In neuroinflammation, distinguishing microglia from macrophages and identifying microglial-specific biomarkers in peripheral blood pose significant challenges. This study comprehensively profiled the extracellular vesicles (EVs) of microglia and macrophages, respectively, revealing co-expressed EVs with UCHL1 and CX3CR1 as EVs derived specifically from microglia in human blood. After extensive validation, using optimized nano flow cytometry, we evaluated plasma CX3CR1+/UCHL1+ EVs across clinical cohorts [multiple sclerosis (MS), HTLV-1 associated myelopathy (HAM), Alzheimer's disease (AD), and Parkinson's disease (PD)], along with established neurodegenerative markers (NMDAR2A and NFL). The findings discovered a notable rise in CX3CR1+/UCHL1+ EVs in MS, particularly heightened in HAM, in contrast to controls. Conversely, AD and PD exhibited unaltered or diminished levels of microglial EVs. An integrated model of CX3CR1+/UCHL1+, NMDAR2A+, and NFL+ EVs demonstrated promising diagnostic potential for distinguishing MS from controls and HAM. As to the disease duration, CX3CR1+/UCHL1+ EVs increased in the initial five years of MS, stabilizing thereafter, whereas NMDAR2A+ and NFL+ EVs remained stable initially but increased significantly in the subsequent five years, suggesting their correlation with disease duration. This study uncovers unique blood microglial EVs with potential as biomarkers for MS diagnosis, differentiation from HAM, and correlation with disease duration.
Collapse
Affiliation(s)
- Jing Duan
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Aowei Lv
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen Guo
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qi Liu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Tian
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ying Yang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jin Bi
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xintong Yu
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Benyan Luo
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhijian Cai
- Institute of Immunology, Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Bin Xu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Ying Fu
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- National Human Brain Bank for Health and Disease, Zhejiang University, Hangzhou, 310012, China.
| |
Collapse
|
4
|
Ji Q, Li Z, Guo Y, Zhang X. S100A9, as a potential predictor of prognosis and immunotherapy response for GBM, promotes the malignant progression of GBM cells and migration of M2 macrophages. Aging (Albany NY) 2024; 16:11513-11534. [PMID: 39137310 PMCID: PMC11346789 DOI: 10.18632/aging.205949] [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: 07/24/2023] [Accepted: 04/22/2024] [Indexed: 08/15/2024]
Abstract
In the past decades, the therapeutic effect of glioblastoma (GBM) has not been significantly improved. Generous evidence indicates that S100A9 has a wide range of functions in tumors, but its exploration in GBM is less. The purpose of this study is to conduct a comprehensive bioinformatics analysis and cytological experiment on S100A9 in GBM. The expression data and clinical data of GBM samples were downloaded from the public database, and comprehensive bioinformatics analysis was performed on S100A9 in GBM using R software. Wound healing assay and transwell assay were used to detect the migration activity of cells, and colony formation assay, EdU staining, and CCK-8 assay were used to detect the proliferation activity of cells. The effect of S100A9 on the migration activity of M2 macrophages was verified by the cell co-culture method. The protein expression was detected by western blotting and immunohistochemical staining. S100A9 is an independent prognostic factor in GBM patients and is related to poor prognosis. It can be used as an effective tool to predict the response of GBM patients to immune checkpoint inhibitors (ICIs). In addition, S100A9 can promote the malignant progression of GBM and the migration of M2 macrophages. On the whole, our study highlights the potential value of S100A9 in predicting prognosis and immunotherapeutic response in GBM patients. More importantly, S100A9 may promote the malignant progress of GBM by involving in some carcinogenic pathways and remodeling the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Qiankun Ji
- Department of Neurosurgery, Zhoukou Central Hospital, Zhoukou, Henan 466000, P.R. China
| | - Zibo Li
- Department of Neurosurgery, Zhoukou Central Hospital, Zhoukou, Henan 466000, P.R. China
| | - Yazhou Guo
- Department of Neurosurgery, Zhoukou Central Hospital, Zhoukou, Henan 466000, P.R. China
| | - Xiaoyang Zhang
- Department of Neurosurgery, Zhoukou Central Hospital, Zhoukou, Henan 466000, P.R. China
| |
Collapse
|
5
|
Zhou Q, Wang Y, Zhang Q, Wei X, Yao Y, Xia L. Noninvasive prediction of CCL2 expression level in high-grade glioma patients. Cancer Med 2024; 13:e70016. [PMID: 39030882 PMCID: PMC11257997 DOI: 10.1002/cam4.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Gliomas are recognized as the most frequent type of malignancies in the central nervous system, and efficacious prognostic indicators are essential to treat patients with gliomas and improve their clinical outcomes. The chemokine (C-C motif) ligand 2 (CCL2) is a promising predictor for glioma malignancy and progression. However, at present, the methods to evaluate CCL2 expression level are invasive and operator-dependent. OBJECTIVE It was expected to noninvasively predict CCL2 expression levels in malignant glioma tissues by magnetic resonance imaging (MRI)-based radiomics and assess the association between the developed radiomics model and prognostic indicators and related genes. METHODS MRI-based radiomics was used to predict CCL2 expression level using data obtained from The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA) databases. A support vector machine (SVM)-based radiomics model and a logistic regression (LR)-based radiomics model were used to predict the radiomics score, and its correlation with CCL2 expression level was analyzed. RESULTS The results revealed that there was an association between CCL2 expression level and the overall survival of cases with gliomas, and bioinformatics correlation analysis showed that CCL2 expression level was highly correlated with disease-related pathways, such as mTOR signaling pathway, cGMP-PKG signaling pathway, and MAPK signaling pathway. Both SVM- and LR-based radiomics data robustly predicted CCL2 expression level, and radiomics scores could also be used to predict the overall survival of patients. Moreover, the high/low radiomics scores were highly correlated with the known glioma-related genes, including CD70, CD27, and PDCD1. CONCLUSION An MRI-based radiomics model was successfully developed, and its clinical benefits were confirmed, including the prediction of CCL2 expression level and patients' prognosis.
Collapse
Affiliation(s)
- Qingqing Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Yamei Wang
- Department of NeurologyThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Qing Zhang
- Department of RadiologyThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - XiaoMing Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Yuan Yao
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Liang Xia
- Department of NeurosurgeryThe Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of SciencesHangzhouPeople's Republic of China
| |
Collapse
|
6
|
Chen Z, Ford KP, Islam MBAR, Wan H, Han H, Ramakrishnan A, Brown RJ, Villanueva V, Wang Y, Davis BT, Weiss C, Cui W, Gate D, Schwulst SJ. antiCD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8+ T-cell cytotoxicity after traumatic brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.596673. [PMID: 38948775 PMCID: PMC11212861 DOI: 10.1101/2024.06.17.596673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people. Graphic abstract Aged brains after TBI comprise two pools of CD8 + T cells . The aged brain has long been resided by a population of CD8 + T cells that's exhaustive and dysfunctional. Post TBI, due to BBB impairment, functional CD8 + T cells primarily migrate into the brain parenchyma. Aged, injury-associated microglia with upregulated MHC class I molecules can present neoantigens such as neuronal and/or myelin debris in the injured brains to functional CD8+ T, resulting in downstream CD8+ T cell cytotoxicity. aCD49d Ab treatment exerts its function by blocking the migration of functional effector CD8 + T cell population, leading to less cytotoxicity and resulting in improved TBI outcomes in aged mice.
Collapse
|
7
|
Koula G, Yakati V, Rachamalla HK, Bhamidipati K, Kathirvel M, Banerjee R, Puvvada N. Integrin receptor-targeted, doxorubicin-loaded cerium oxide nanoparticles delivery to combat glioblastoma. Nanomedicine (Lond) 2024; 19:1389-1406. [PMID: 38912661 PMCID: PMC11318704 DOI: 10.1080/17435889.2024.2350357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/29/2024] [Indexed: 06/25/2024] Open
Abstract
Aim: To assess the chemo-immunomodulatory effects of doxorubicin-loaded cerium oxide nanoparticles coated with oleyl amine-linked cyclic RGDfK peptide (CeNP+Dox+RGD) to target both gliomas and its tumor microenvironment (TME) via integrin receptors. Materials & methods: CeNP+Dox+RGD nanoparticles are synthesized by the sequential addition of cerium III chloride heptahydrate, beta-cyclodextrin, oleic acid, and F127 micelle (CeNP). Doxorubicin was then loaded into CeNPs and coated with oleyl amine-linked cyclic RGDfK peptide to form stable CeNP+Dox+RGD nanoparticles. Results: CeNP+Dox+RGD nanoparticles crossed blood-brain barrier (BBB) effectively and demonstrated threefold enhanced survivability in glioma-bearing mice. The IHC profiling of glial tumor cross-sections showed increased CD80 expression (M1 TAMs) and decreased arginase-1 expression (M2 TAMs). Conclusion: CeNP+Dox+RGD can be an immunotherapeutic treatment option to combat glioblastoma.
Collapse
Affiliation(s)
- Gayathri Koula
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Venu Yakati
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Hari Krishnareddy Rachamalla
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Keerti Bhamidipati
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh (U.P), India
| | - Muralidharan Kathirvel
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Rajkumar Banerjee
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Nagaprasad Puvvada
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| |
Collapse
|
8
|
Yabo YA, Moreno-Sanchez PM, Pires-Afonso Y, Kaoma T, Nosirov B, Scafidi A, Ermini L, Lipsa A, Oudin A, Kyriakis D, Grzyb K, Poovathingal SK, Poli A, Muller A, Toth R, Klink B, Berchem G, Berthold C, Hertel F, Mittelbronn M, Heiland DH, Skupin A, Nazarov PV, Niclou SP, Michelucci A, Golebiewska A. Glioblastoma-instructed microglia transition to heterogeneous phenotypic states with phagocytic and dendritic cell-like features in patient tumors and patient-derived orthotopic xenografts. Genome Med 2024; 16:51. [PMID: 38566128 PMCID: PMC10988817 DOI: 10.1186/s13073-024-01321-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. METHODS Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry, and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. RESULTS We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood-brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. CONCLUSIONS Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.
Collapse
Affiliation(s)
- Yahaya A Yabo
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
| | - Pilar M Moreno-Sanchez
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
| | - Yolanda Pires-Afonso
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1210, Luxembourg, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Bakhtiyor Nosirov
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Andrea Scafidi
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1210, Luxembourg, Luxembourg
| | - Luca Ermini
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg
| | - Anuja Lipsa
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg
| | - Dimitrios Kyriakis
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Suresh K Poovathingal
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Single Cell Analytics & Microfluidics Core, Vlaams Instituut Voor Biotechnologie-KU Leuven, 3000, Louvain, Belgium
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1210, Luxembourg, Luxembourg
| | - Arnaud Muller
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Reka Toth
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Barbara Klink
- National Center of Genetics, Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
- Department of Cancer Research, Luxembourg Institute of Health, L-1210, Luxembourg, Luxembourg
- German Cancer Consortium (DKTK): Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Cancer Consortium (DKTK) Partner Site Dresden, and German Cancer Research Center (DKFZ), Dresden, Heidelberg, 01307, Germany
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Guy Berchem
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
- Department of Cancer Research, Luxembourg Institute of Health, L-1210, Luxembourg, Luxembourg
- Centre Hospitalier Luxembourg, L-1210, Luxembourg, Luxembourg
| | | | - Frank Hertel
- Centre Hospitalier Luxembourg, L-1210, Luxembourg, Luxembourg
| | - Michel Mittelbronn
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Cancer Research, Luxembourg Institute of Health, L-1210, Luxembourg, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
| | - Dieter H Heiland
- Translational Neurosurgery, Friedrich-Alexander University Erlangen Nuremberg, 91054, Erlangen, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, 91054, Erlangen, Germany
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Neurosurgery, Medical Center, University of Freiburg, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, 79106, Freiburg, Germany
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Physics and Material Science, University Luxembourg, L-4367, Belvaux, Luxembourg
- Department of Neuroscience, University of California San Diego, La Jolla, CA, 92093, USA
| | - Petr V Nazarov
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367, Belvaux, Luxembourg
| | - Alessandro Michelucci
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg.
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1210, Luxembourg, Luxembourg.
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg.
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210, Luxembourg, Luxembourg.
| |
Collapse
|
9
|
Zhang SS, Li RQ, Chen Z, Wang XY, Dumont AS, Fan X. Immune cells: potential carriers or agents for drug delivery to the central nervous system. Mil Med Res 2024; 11:19. [PMID: 38549161 PMCID: PMC10979586 DOI: 10.1186/s40779-024-00521-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
Drug delivery systems (DDS) have recently emerged as a promising approach for the unique advantages of drug protection and targeted delivery. However, the access of nanoparticles/drugs to the central nervous system (CNS) remains a challenge mainly due to the obstruction from brain barriers. Immune cells infiltrating the CNS in the pathological state have inspired the development of strategies for CNS foundation drug delivery. Herein, we outline the three major brain barriers in the CNS and the mechanisms by which immune cells migrate across the blood-brain barrier. We subsequently review biomimetic strategies utilizing immune cell-based nanoparticles for the delivery of nanoparticles/drugs to the CNS, as well as recent progress in rationally engineering immune cell-based DDS for CNS diseases. Finally, we discuss the challenges and opportunities of immune cell-based DDS in CNS diseases to promote their clinical development.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Ruo-Qi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Zhong Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Xiao-Ying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA.
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
10
|
Chia TY, Billingham LK, Boland L, Katz JL, Arrieta VA, Shireman J, Rosas AL, DeLay SL, Zillinger K, Geng Y, Kruger J, Silvers C, Wang H, Vazquez Cervantes GI, Hou D, Wang S, Wan H, Sonabend A, Zhang P, Lee-Chang C, Miska J. The CXCL16-CXCR6 axis in glioblastoma modulates T-cell activity in a spatiotemporal context. Front Immunol 2024; 14:1331287. [PMID: 38299146 PMCID: PMC10827847 DOI: 10.3389/fimmu.2023.1331287] [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: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction Glioblastoma multiforme (GBM) pathobiology is characterized by its significant induction of immunosuppression within the tumor microenvironment, predominantly mediated by immunosuppressive tumor-associated myeloid cells (TAMCs). Myeloid cells play a pivotal role in shaping the GBM microenvironment and influencing immune responses, with direct interactions with effector immune cells critically impacting these processes. Methods Our study investigates the role of the CXCR6/CXCL16 axis in T-cell myeloid interactions within GBM tissues. We examined the surface expression of CXCL16, revealing its limitation to TAMCs, while microglia release CXCL16 as a cytokine. The study explores how these distinct expression patterns affect T-cell engagement, focusing on the consequences for T-cell function within the tumor environment. Additionally, we assessed the significance of CXCR6 expression in T-cell activation and the initial migration to tumor tissues. Results Our data demonstrates that CXCL16 surface expression on TAMCs results in predominant T-cell engagement with these cells, leading to impaired T-cell function within the tumor environment. Conversely, our findings highlight the essential role of CXCR6 expression in facilitating T-cell activation and initial migration to tumor tissues. The CXCL16-CXCR6 axis exhibits dualistic characteristics, facilitating the early stages of the T-cell immune response and promoting T-cell infiltration into tumors. However, once inside the tumor, this axis contributes to immunosuppression. Discussion The dual nature of the CXCL16-CXCR6 axis underscores its potential as a therapeutic target in GBM. However, our results emphasize the importance of carefully considering the timing and context of intervention. While targeting this axis holds promise in combating GBM, the complex interplay between TAMCs, microglia, and T cells suggests that intervention strategies need to be tailored to optimize the balance between promoting antitumor immunity and preventing immunosuppression within the dynamic tumor microenvironment.
Collapse
Affiliation(s)
- Tzu-Yi Chia
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Leah K. Billingham
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lauren Boland
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, United States
| | - Joshua L. Katz
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Victor A. Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jack Shireman
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI, United States
| | - Aurora-Lopez Rosas
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Susan L. DeLay
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kaylee Zillinger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Yuheng Geng
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jeandre Kruger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Caylee Silvers
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hanxiang Wang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Gustavo Ignacio Vazquez Cervantes
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - David Hou
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Si Wang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hanxiao Wan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Adam Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Peng Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
11
|
DePaula-Silva AB. The Contribution of Microglia and Brain-Infiltrating Macrophages to the Pathogenesis of Neuroinflammatory and Neurodegenerative Diseases during TMEV Infection of the Central Nervous System. Viruses 2024; 16:119. [PMID: 38257819 PMCID: PMC10819099 DOI: 10.3390/v16010119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The infection of the central nervous system (CNS) with neurotropic viruses induces neuroinflammation and is associated with the development of neuroinflammatory and neurodegenerative diseases, including multiple sclerosis and epilepsy. The activation of the innate and adaptive immune response, including microglial, macrophages, and T and B cells, while required for efficient viral control within the CNS, is also associated with neuropathology. Under healthy conditions, resident microglia play a pivotal role in maintaining CNS homeostasis. However, during pathological events, such as CNS viral infection, microglia become reactive, and immune cells from the periphery infiltrate into the brain, disrupting CNS homeostasis and contributing to disease development. Theiler's murine encephalomyelitis virus (TMEV), a neurotropic picornavirus, is used in two distinct mouse models: TMEV-induced demyelination disease (TMEV-IDD) and TMEV-induced seizures, representing mouse models of multiple sclerosis and epilepsy, respectively. These murine models have contributed substantially to our understanding of the pathophysiology of MS and seizures/epilepsy following viral infection, serving as critical tools for identifying pharmacological targetable pathways to modulate disease development. This review aims to discuss the host-pathogen interaction during a neurotropic picornavirus infection and to shed light on our current understanding of the multifaceted roles played by microglia and macrophages in the context of these two complexes viral-induced disease.
Collapse
Affiliation(s)
- Ana Beatriz DePaula-Silva
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
12
|
Nafe R, Hattingen E. Cellular Components of the Tumor Environment in Gliomas-What Do We Know Today? Biomedicines 2023; 12:14. [PMID: 38275375 PMCID: PMC10813739 DOI: 10.3390/biomedicines12010014] [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: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
A generation ago, the molecular properties of tumor cells were the focus of scientific interest in oncology research. Since then, it has become increasingly apparent that the tumor environment (TEM), whose major components are non-neoplastic cell types, is also of utmost importance for our understanding of tumor growth, maintenance and resistance. In this review, we present the current knowledge concerning all cellular components within the TEM in gliomas, focusing on their molecular properties, expression patterns and influence on the biological behavior of gliomas. Insight into the TEM of gliomas has expanded considerably in recent years, including many aspects that previously received only marginal attention, such as the phenomenon of phagocytosis of glioma cells by macrophages and the role of the thyroid-stimulating hormone on glioma growth. We also discuss other topics such as the migration of lymphocytes into the tumor, phenotypic similarities between chemoresistant glioma cells and stem cells, and new clinical approaches with immunotherapies involving the cells of TEM.
Collapse
Affiliation(s)
- Reinhold Nafe
- Department of Neuroradiology, Clinics of Johann Wolfgang Goethe-University, Schleusenweg 2-16, D-60528 Frankfurt am Main, Germany;
| | | |
Collapse
|
13
|
Yabo YA, Moreno-Sanchez PM, Pires-Afonso Y, Kaoma T, Nosirov B, Scafidi A, Ermini L, Lipsa A, Oudin A, Kyriakis D, Grzyb K, Poovathingal SK, Poli A, Muller A, Toth R, Klink B, Berchem G, Berthold C, Hertel F, Mittelbronn M, Heiland DH, Skupin A, Nazarov PV, Niclou SP, Michelucci A, Golebiewska A. Glioblastoma-instructed microglia transition to heterogeneous phenotypic states with phagocytic and dendritic cell-like features in patient tumors and patient-derived orthotopic xenografts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.05.531162. [PMID: 36945572 PMCID: PMC10028830 DOI: 10.1101/2023.03.05.531162] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Background A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. Methods Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA-sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. Results We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood-brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. Conclusions Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.
Collapse
Affiliation(s)
- Yahaya A Yabo
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Pilar M Moreno-Sanchez
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Yolanda Pires-Afonso
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Tony Kaoma
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Bakhtiyor Nosirov
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Andrea Scafidi
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Luca Ermini
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
| | - Anuja Lipsa
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
| | - Dimitrios Kyriakis
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Suresh K Poovathingal
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- Single Cell Analytics & Microfluidics Core, Vlaams Instituut voor Biotechnologie-KU Leuven, 3000 Leuven, Belgium
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Arnaud Muller
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Reka Toth
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Barbara Klink
- National Center of Genetics, Laboratoire National de Santé, L-3555 Dudelange, Luxembourg
- Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
- German Cancer Consortium (DKTK), 01307 Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), 01307 Dresden, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Guy Berchem
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
- Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
- Centre Hospitalier Luxembourg, 1210 Luxembourg, Luxembourg
| | | | - Frank Hertel
- Centre Hospitalier Luxembourg, 1210 Luxembourg, Luxembourg
| | - Michel Mittelbronn
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé, L-3555 Dudelange, Luxembourg
| | - Dieter H Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center - University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- Department of Physics and Material Science, University Luxembourg, L-4367 Belvaux, Luxembourg
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92093, USA
| | - Petr V Nazarov
- Multiomics Data Science, Department of Cancer Research, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Alessandro Michelucci
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg, Luxembourg
| |
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
|
Rabah N, Ait Mohand FE, Kravchenko-Balasha N. Understanding Glioblastoma Signaling, Heterogeneity, Invasiveness, and Drug Delivery Barriers. Int J Mol Sci 2023; 24:14256. [PMID: 37762559 PMCID: PMC10532387 DOI: 10.3390/ijms241814256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The most prevalent and aggressive type of brain cancer, namely, glioblastoma (GBM), is characterized by intra- and inter-tumor heterogeneity and strong spreading capacity, which makes treatment ineffective. A true therapeutic answer is still in its infancy despite various studies that have made significant progress toward understanding the mechanisms behind GBM recurrence and its resistance. The primary causes of GBM recurrence are attributed to the heterogeneity and diffusive nature; therefore, monitoring the tumor's heterogeneity and spreading may offer a set of therapeutic targets that could improve the clinical management of GBM and prevent tumor relapse. Additionally, the blood-brain barrier (BBB)-related poor drug delivery that prevents effective drug concentrations within the tumor is discussed. With a primary emphasis on signaling heterogeneity, tumor infiltration, and computational modeling of GBM, this review covers typical therapeutic difficulties and factors contributing to drug resistance development and discusses potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem 91120, Israel; (N.R.); (F.-E.A.M.)
| |
Collapse
|
16
|
Scieszka D, Jin Y, Noor S, Barr E, Garcia M, Begay J, Herbert G, Hunter RP, Bhaskar K, Kumar R, Gullapalli R, Bolt A, McCormick MA, Bleske B, Gu H, Campen MJ. Biomass smoke inhalation promotes neuroinflammatory and metabolomic temporal changes in the hippocampus of female mice. J Neuroinflammation 2023; 20:192. [PMID: 37608305 PMCID: PMC10464132 DOI: 10.1186/s12974-023-02874-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/15/2023] [Indexed: 08/24/2023] Open
Abstract
Smoke from wildland fires has been shown to produce neuroinflammation in preclinical models, characterized by neural infiltrations of neutrophils and monocytes, as well as altered neurovascular endothelial phenotypes. To address the longevity of such outcomes, the present study examined the temporal dynamics of neuroinflammation and metabolomics after inhalation exposures from biomass-derived smoke. 2-month-old female C57BL/6 J mice were exposed to wood smoke every other day for 2 weeks at an average exposure concentration of 0.5 mg/m3. Subsequent serial euthanasia occurred at 1-, 3-, 7-, 14-, and 28-day post-exposure. Flow cytometry of right hemispheres revealed two endothelial populations of CD31Hi and CD31Med expressors, with wood smoke inhalation causing an increased proportion of CD31Hi. These populations of CD31Hi and CD31Med were associated with an anti-inflammatory and pro-inflammatory response, respectively, and their inflammatory profiles were largely resolved by the 28-day mark. However, activated microglial populations (CD11b+/CD45low) remained higher in wood smoke-exposed mice than controls at day 28. Infiltrating neutrophil populations decreased to levels below controls by day 28. However, the MHC-II expression of the peripheral immune infiltrate remained high, and the population of neutrophils retained an increased expression of CD45, Ly6C, and MHC-II. Utilizing an unbiased approach examining the metabolomic alterations, we observed notable hippocampal perturbations in neurotransmitter and signaling molecules, such as glutamate, quinolinic acid, and 5-α-dihydroprogesterone. Utilizing a targeted panel designed to explore the aging-associated NAD+ metabolic pathway, wood smoke exposure drove fluctuations and compensations across the 28-day time course, ending with decreased hippocampal NAD+ abundance on day 28. Summarily, these results indicate a highly dynamic neuroinflammatory environment, with potential resolution extending past 28 days, the implications of which may include long-term behavioral changes, systemic and neurological sequalae directly associated with wildfire smoke exposure.
Collapse
Affiliation(s)
- David Scieszka
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Yan Jin
- Florida International University Center for Translational Sciences, Port St. Lucie, FL, 34987, USA
| | - Shahani Noor
- Department of Molecular Genetics and Microbiology, Department of Neurology, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Ed Barr
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Marcus Garcia
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Jessica Begay
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Guy Herbert
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Russell P Hunter
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, Department of Neurology, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Rahul Kumar
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Rama Gullapalli
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Alicia Bolt
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Mark A McCormick
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Barry Bleske
- Department of Pharmacy Practice and Administrative Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Haiwei Gu
- Florida International University Center for Translational Sciences, Port St. Lucie, FL, 34987, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA.
| |
Collapse
|
17
|
Tai SH, Chao LC, Huang SY, Lin HW, Lee AH, Chen YY, Lee EJ. Nicotinamide Deteriorates Post-Stroke Immunodepression Following Cerebral Ischemia-Reperfusion Injury in Mice. Biomedicines 2023; 11:2145. [PMID: 37626642 PMCID: PMC10452067 DOI: 10.3390/biomedicines11082145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Inducing experimental stroke leads to biphasic immune responses, where the early activation of immune functions is followed by severe immunosuppression accompanied by spleen and thymus atrophy. Nicotinamide, a water-soluble B-group vitamin, is a known neuroprotectant against brain ischemia in animal models. We examined the effect of nicotinamide on the central and peripheral immune response in experimental stroke models. (2) Methods: Nicotinamide (500 mg/kg) or saline was intravenously administered to C57BL/6 mice during reperfusion after transiently occluding the middle cerebral artery or after LPS injection. On day 3, the animals were examined for behavioral performance and were then sacrificed to assess brain infarction, blood-brain barrier (BBB) integrity, and the composition of immune cells in the brain, thymus, spleen, and blood using flow cytometry. (3) Results: Nicotinamide reduced brain infarction and microglia/macrophage activation following MCAo (p < 0.05). Similarly, in LPS-injected mice, microglia/macrophage activation was decreased upon treatment with nicotinamide (p < 0.05), suggesting a direct inhibitory effect of nicotinamide on microglia/macrophage activation. Nicotinamide decreased the infiltration of neutrophils into the brain parenchyma and ameliorated Evans blue leakage (p < 0.05), suggesting that a decreased infiltration of neutrophils could, at least partially, be the result of a more integrated BBB structure following nicotinamide treatment. Our studies also revealed that administering nicotinamide led to retarded B-cell maturation in the spleen and subsequently decreased circulating B cells in the thymus and bloodstream (p < 0.05). (4) Conclusions: Cumulatively, nicotinamide decreased brain inflammation caused by ischemia-reperfusion injury, which was mediated by a direct anti-inflammatory effect of nicotinamide and an indirect protective effect on BBB integrity. Administering nicotinamide following brain ischemia resulted in a decrease in circulating B cells. This warrants attention with respect to future clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - E-Jian Lee
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| |
Collapse
|
18
|
Scieszka D, Jin Y, Noor S, Barr E, Garcia M, Begay J, Herbert G, Hunter RP, Bhaskar K, Kumar R, Gullapalli R, Bolt A, McCormick MA, Bleske B, Gu H, Campen M. Neuroinflammatory and Metabolomic Temporal Dynamics Following Wood Smoke Inhalation. RESEARCH SQUARE 2023:rs.3.rs-3002040. [PMID: 37333410 PMCID: PMC10275049 DOI: 10.21203/rs.3.rs-3002040/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Smoke from wildland fires has been shown to produce neuroinflammation in preclinical models, characterized by neural infiltrations of neutrophils and monocytes, as well as altered neurovascular endothelial phenotypes. To address the longevity of such outcomes, the present study examined the neuroinflammatory and metabolomic temporal dynamics after inhalation exposures from biomass-derived smoke. 2-month-old female C57BL/6J mice were exposed to wood smoke every other day for two weeks at an average exposure concentration of 0.5mg/m 3 . Subsequent serial euthanasia occurred at 1-, 3-, 7-, 14-, and 28-days post-exposure. Flow cytometry of right hemispheres revealed two endothelial populations of PECAM (CD31), high and medium expressors, with wood smoke inhalation causing an increased proportion of PECAM Hi . These populations of PECAM Hi and PECAM Med were associated with an anti-inflammatory and pro-inflammatory response, respectively, and their inflammatory profiles were largely resolved by the 28-day mark. However, activated microglial populations (CD11b + /CD45 low ) remained higher in wood smoke-exposed mice than controls at day 28. Infiltrating neutrophil populations decreased to levels below controls by day 28. However, the MHC-II expression of the peripheral immune infiltrate remained high, and the population of neutrophils retained an increased expression of CD45, Ly6C, and MHC-II. Utilizing an unbiased approach examining the metabolomic alterations, we observed notable hippocampal perturbations in neurotransmitter and signaling molecules like glutamate, quinolinic acid, and 5-α-dihydroprogesterone. Utilizing a targeted panel designed to explore the aging-associated NAD + metabolic pathway, wood smoke exposure drove fluctuations and compensations across the 28-day time course, ending with decreased hippocampal NAD + abundance at day 28. Summarily, these results indicate a highly dynamic neuroinflammatory environment, with potential resolution extending past 28 days, the implications of which may include long-term behavioral changes, systemic and neurological sequalae directly associated wtith wildfire smoke exposure.
Collapse
Affiliation(s)
| | - Yan Jin
- Florida International University, Center for Translational Sciences
| | - Shahani Noor
- University of New Mexico, Department of Molecular Genetics and Microbiology
| | - Ed Barr
- University of New Mexico, College of Pharmacy
| | | | | | - Guy Herbert
- University of New Mexico, College of Pharmacy
| | | | - Kiran Bhaskar
- University of New Mexico, Department of Molecular Genetics and Microbiology
| | - Rahul Kumar
- University of New Mexico, Department of Pathology
| | | | - Alicia Bolt
- University of New Mexico, College of Pharmacy
| | - Mark A McCormick
- University of New Mexico, Department of Biochemistry and Molecular Biology
| | - Barry Bleske
- University of New Mexico, Department of Pharmacy Practice and Administrative Science
| | - Haiwei Gu
- Florida International University, Center for Translational Sciences
| | | |
Collapse
|
19
|
Montemurro N, Pahwa B, Tayal A, Shukla A, De Jesus Encarnacion M, Ramirez I, Nurmukhametov R, Chavda V, De Carlo A. Macrophages in Recurrent Glioblastoma as a Prognostic Factor in the Synergistic System of the Tumor Microenvironment. Neurol Int 2023; 15:595-608. [PMID: 37218976 DOI: 10.3390/neurolint15020037] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM) is a common and highly malignant primary tumor of the central nervous system in adults. Ever more recent papers are focusing on understanding the role of the tumor microenvironment (TME) in affecting tumorigenesis and the subsequent prognosis. We assessed the impact of macrophages in the TME on the prognosis in patients with recurrent GBM. A PubMed, MEDLINE and Scopus review was conducted to identify all studies dealing with macrophages in the GBM microenvironment from January 2016 to December 2022. Glioma-associated macrophages (GAMs) act critically in enhancing tumor progression and can alter drug resistance, promoting resistance to radiotherapy and establishing an immunosuppressive environment. M1 macrophages are characterized by increased secretion of proinflammatory cytokines, such as IL-1ß, tumor necrosis factor (TNF), IL-27, matrix metalloproteinase (MMPs), CCL2, and VEGF (vascular endothelial growth factor), IGF1, that can lead to the destruction of the tissue. In contrast, M2 is supposed to participate in immunosuppression and tumor progression, which is formed after being exposed to the macrophage M-CSF, IL-10, IL-35 and the transforming growth factor-ß (TGF-β). Because there is currently no standard of care in recurrent GBM, novel identified targeted therapies based on the complex signaling and interactions between the glioma stem cells (GSCs) and the TME, especially resident microglia and bone-marrow-derived macrophages, may be helpful in improving the overall survival of these patients in the near future.
Collapse
Affiliation(s)
- Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| | - Bhavya Pahwa
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anish Tayal
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anushruti Shukla
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | | | - Issael Ramirez
- Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Renat Nurmukhametov
- Department of Spinal Surgery, Central Clinical Hospital of the Russian Academy of Sciences, 121359 Moscow, Russia
| | - Vishal Chavda
- Department of Pathology, Stanford of School of Medicine, Stanford University Medical Centre, Palo Alto, CA 94305, USA
| | - Antonella De Carlo
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| |
Collapse
|
20
|
Identification and Characterization of TMEM119-Positive Cells in the Postnatal and Adult Murine Cochlea. Brain Sci 2023; 13:brainsci13030516. [PMID: 36979326 PMCID: PMC10046579 DOI: 10.3390/brainsci13030516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/09/2023] [Accepted: 03/18/2023] [Indexed: 03/22/2023] Open
Abstract
Transmembrane protein 119 (TMEM119) is expressed in a subset of resident macrophage cells of the brain and was proposed as a marker for native brain microglia. The presence of cells expressing TMEM119 in the cochlea has not yet been described. Thus, the present study aimed to characterize the TMEM119-expressing cells of the postnatal and adult cochlea, the latter also after noise exposure. Immunofluorescent staining of cochlear cryosections detected TMEM119 protein in the spiral limbus fibrocytes and the developing stria vascularis at postnatal Day 3. Applying the macrophage marker Iba1 revealed that TMEM119 is not a marker of cochlear macrophages or a subset of them. In the adult murine cochlea, TMEM119 expression was detected in the basal cells of the stria vascularis and the dark mesenchymal cells of the supralimbal zone. Exposure to noise trauma was not associated with a qualitative change in the types or distributions of the TMEM119-expressing cells of the adult cochlea. Western blot analysis indicated a similar TMEM119 protein expression level in the postnatal cochlea and brain tissues. The findings do not support using TMEM119 as a specific microglial or macrophage marker in the cochlea. The precise role of TMEM119 in the cochlea remains to be investigated through functional experiments. TMEM119 expression in the basal cells of the stria vascularis implies a possible role in the gap junction system of the blood–labyrinth barrier and merits further research.
Collapse
|
21
|
Glial roles in sterile inflammation after ischemic stroke. Neurosci Res 2023; 187:67-71. [PMID: 36206952 DOI: 10.1016/j.neures.2022.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
Abstract
Stroke is a leading cause of death and disability worldwide, but there are a limited number of therapies that improve patients' functional recovery. The complicated mechanisms of post-stroke neuroinflammation, which is responsible for secondary ischemic neuronal damage, have been clarified by extensive research. Activation of microglia and astrocytes due to ischemic insults is implicated in the production of pro-inflammatory factors, formation of the glial scar, and breakdown of the blood-brain barrier. This leads to the infiltration of leukocytes, which are activated by damage-associated molecular patterns (DAMPs) to produce pro-inflammatory factors and induce additional neuronal damage. In this review, we focus on the glial mechanisms underlying sterile post-ischemic inflammation after stroke.
Collapse
|
22
|
Bungert AD, Urbantat RM, Jelgersma C, Bekele BM, Mueller S, Mueller A, Felsenstein M, Dusatko S, Blank A, Ghori A, Boehm-Sturm P, Koch SP, Vajkoczy P, Brandenburg S. Myeloid cell subpopulations compensate each other for Ccr2-deficiency in glioblastoma. Neuropathol Appl Neurobiol 2023; 49:e12863. [PMID: 36346010 DOI: 10.1111/nan.12863] [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: 11/13/2021] [Revised: 08/07/2022] [Accepted: 10/09/2022] [Indexed: 11/11/2022]
Abstract
AIMS Glioblastomas are high-grade brain tumours that are characterised by the accumulation of brain-resident microglia and peripheral macrophages. Recruitment of these myeloid cells can be facilitated by CCR2/CCL2 signalling. Besides the well-known CCR2+ macrophages, we have identified microglia expressing CCR2 in glioma tissues. Thus, we investigated how Ccr2-deficiency of one of the myeloid cell populations affects the other population and tumour biology. METHODS We generated four chimeric groups to analyse single and combined Ccr2-deficiency of microglia and macrophages. On day 21 after tumour cell implantation (GL261), we conducted flow cytometry, immunofluorescence and real-time polymerase chain reaction analyses. Tumour volume and metabolism were determined by magnetic resonance imaging and magnetic resonance spectroscopy. Moreover, in vitro studies were performed with primary microglia and bone marrow-derived macrophages. RESULTS We demonstrated reduced infiltration of macrophages and microglia depending on the lack of Ccr2. However, the total number of myeloid cells remained constant except for the animals with dual Ccr2-knockout. Both microglia and macrophages with Ccr2-deficiency showed impaired expression of proinflammatory molecules and altered phagocytic activity. Despite the altered immunologic phenotype caused by Ccr2-deficiency, glioma progression and metabolism were hardly affected. Alterations were detected solely in apoptosis and proliferation of tumours from animals with specific Ccr2-deficient microglia, whereas vessel stability was increased in mice with Ccr2-knockout in both cell populations. CONCLUSION These results indicate that microglia and macrophages provide a homoeostatic balance within glioma tissue and compensate for the lack of the corresponding counterpart. Moreover, we identified that the CCR2/CCL2 axis is involved in the immunologic function of microglia and macrophages beyond its relevance for migration.
Collapse
Affiliation(s)
- Alexander D Bungert
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ruth M Urbantat
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claudius Jelgersma
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Biniam M Bekele
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Berlin, Germany
| | - Annett Mueller
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthäus Felsenstein
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Silke Dusatko
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anne Blank
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Adnan Ghori
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Berlin, Germany
| | - Stefan P Koch
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter Vajkoczy
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susan Brandenburg
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
23
|
Corraliza-Gomez M, Bendito B, Sandonis-Camarero D, Mondejar-Duran J, Villa M, Poncela M, Valero J, Sanchez D, Ganfornina MD. Dual role of Apolipoprotein D as long-term instructive factor and acute signal conditioning microglial secretory and phagocytic responses. Front Cell Neurosci 2023; 17:1112930. [PMID: 36779011 PMCID: PMC9908747 DOI: 10.3389/fncel.2023.1112930] [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: 11/30/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Microglial cells are recognized as very dynamic brain cells, screening the environment and sensitive to signals from all other cell types in health and disease. Apolipoprotein D (ApoD), a lipid-binding protein of the Lipocalin family, is required for nervous system optimal function and proper development and maintenance of key neural structures. ApoD has a cell and state-dependent expression in the healthy nervous system, and increases its expression upon aging, damage or neurodegeneration. An extensive overlap exists between processes where ApoD is involved and those where microglia have an active role. However, no study has analyzed the role of ApoD in microglial responses. In this work, we test the hypothesis that ApoD, as an extracellular signal, participates in the intercellular crosstalk sensed by microglia and impacts their responses upon physiological aging or damaging conditions. We find that a significant proportion of ApoD-dependent aging transcriptome are microglia-specific genes, and show that lack of ApoD in vivo dysregulates microglial density in mouse hippocampus in an age-dependent manner. Murine BV2 and primary microglia do not express ApoD, but it can be internalized and targeted to lysosomes, where unlike other cell types it is transiently present. Cytokine secretion profiles and myelin phagocytosis reveal that ApoD has both long-term pre-conditioning effects on microglia as well as acute effects on these microglial immune functions, without significant modification of cell survival. ApoD-triggered cytokine signatures are stimuli (paraquat vs. Aβ oligomers) and sex-dependent. Acute exposure to ApoD induces microglia to switch from their resting state to a secretory and less phagocytic phenotype, while long-term absence of ApoD leads to attenuated cytokine induction and increased myelin uptake, supporting a role for ApoD as priming or immune training factor. This knowledge should help to advance our understanding of the complex responses of microglia during aging and neurodegeneration, where signals received along our lifespan are combined with damage-triggered acute signals, conditioning both beneficial roles and limitations of microglial functions.
Collapse
Affiliation(s)
- Miriam Corraliza-Gomez
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain
| | - Beatriz Bendito
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain
| | - David Sandonis-Camarero
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain
| | - Jorge Mondejar-Duran
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain
| | - Miguel Villa
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain
| | - Marta Poncela
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain
| | - Jorge Valero
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | - Diego Sanchez
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain,Diego Sanchez,
| | - Maria D. Ganfornina
- Instituto de Biología y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid, Spain,*Correspondence: Maria D. Ganfornina, ,
| |
Collapse
|
24
|
The Tumor Immune Microenvironment in Primary CNS Neoplasms: A Review of Current Knowledge and Therapeutic Approaches. Int J Mol Sci 2023; 24:ijms24032020. [PMID: 36768342 PMCID: PMC9917056 DOI: 10.3390/ijms24032020] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Primary CNS neoplasms are responsible for considerable mortality and morbidity, and many therapies directed at primary brain tumors have proven unsuccessful despite their success in preclinical studies. Recently, the tumor immune microenvironment has emerged as a critical aspect of primary CNS neoplasms that may affect their malignancy, prognosis, and response to therapy across patients and tumor grades. This review covers the tumor microenvironment of various primary CNS neoplasms, with a focus on glioblastoma and meningioma. Additionally, current therapeutic strategies based on elements of the tumor microenvironment, including checkpoint inhibitor therapy and immunotherapeutic vaccines, are discussed.
Collapse
|
25
|
Myeloid cell heterogeneity in the tumor microenvironment and therapeutic implications for childhood central nervous system (CNS) tumors. J Neuroimmunol 2023; 374:578009. [PMID: 36508930 DOI: 10.1016/j.jneuroim.2022.578009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/07/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022]
Abstract
Central nervous system (CNS) tumors are the most common type of solid tumors in children and the leading cause of cancer deaths in ages 0-14. Recent advances in the field of tumor biology and immunology have underscored the disparate nature of these distinct CNS tumor types. In this review, we briefly introduce pediatric CNS tumors and discuss various components of the TME, with a particular focus on myeloid cells. Although most studies regarding myeloid cells have been done on adult CNS tumors and animal models, we discuss the role of myeloid cell heterogeneity in pediatric CNS tumors and describe how these cells may contribute to tumorigenesis and treatment response. In addition, we present studies within the last 5 years that highlight human CNS tumors, the utility of various murine CNS tumor models, and the latest multi-dimensional tools that can be leveraged to investigate myeloid cell infiltration in young adults and children diagnosed with select CNS tumors.
Collapse
|
26
|
Khan F, Pang L, Dunterman M, Lesniak MS, Heimberger AB, Chen P. Macrophages and microglia in glioblastoma: heterogeneity, plasticity, and therapy. J Clin Invest 2023; 133:163446. [PMID: 36594466 PMCID: PMC9797335 DOI: 10.1172/jci163446] [Citation(s) in RCA: 106] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive tumor in the central nervous system and contains a highly immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages and microglia (TAMs) are a dominant population of immune cells in the GBM TME that contribute to most GBM hallmarks, including immunosuppression. The understanding of TAMs in GBM has been limited by the lack of powerful tools to characterize them. However, recent progress on single-cell technologies offers an opportunity to precisely characterize TAMs at the single-cell level and identify new TAM subpopulations with specific tumor-modulatory functions in GBM. In this Review, we discuss TAM heterogeneity and plasticity in the TME and summarize current TAM-targeted therapeutic potential in GBM. We anticipate that the use of single-cell technologies followed by functional studies will accelerate the development of novel and effective TAM-targeted therapeutics for GBM patients.
Collapse
|
27
|
Pachocki CJ, Hol EM. Current perspectives on diffuse midline glioma and a different role for the immune microenvironment compared to glioblastoma. J Neuroinflammation 2022; 19:276. [PMCID: PMC9675250 DOI: 10.1186/s12974-022-02630-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022] Open
Abstract
Diffuse midline glioma (DMG), formerly called diffuse intrinsic pontine glioma (DIPG), is a high-grade malignant pediatric brain tumor with a near-zero survival rate. To date, only radiation therapy provides marginal survival benefit; however, the median survival time remains less than a year. Historically, the infiltrative nature and sensitive location of the tumor rendered surgical removal and biopsies difficult and subsequently resulted in limited knowledge of the disease, as only post-mortem tissue was available. Therefore, clinical decision-making was based upon experience with the more frequent and histologically similar adult glioblastoma (GBM). Recent advances in tissue acquisition and molecular profiling revealed that DMG and GBM are distinct disease entities, with separate tissue characteristics and genetic profiles. DMG is characterized by heterogeneous tumor tissue often paired with an intact blood–brain barrier, possibly explaining its resistance to chemotherapy. Additional profiling shed a light on the origin of the disease and the influence of several mutations such as a highly recurring K27M mutation in histone H3 on its tumorigenesis. Furthermore, early evidence suggests that DMG has a unique immune microenvironment, characterized by low levels of immune cell infiltration, inflammation, and immunosuppression that may impact disease development and outcome. Within the tumor microenvironment of GBM, tumor-associated microglia/macrophages (TAMs) play a large role in tumor development. Interestingly, TAMs in DMG display distinct features and have low immune activation in comparison to other pediatric gliomas. Although TAMs have been investigated substantially in GBM over the last years, this has not been the case for DMG due to the lack of tissue for research. Bit by bit, studies are exploring the TAM–glioma crosstalk to identify what factors within the DMG microenvironment play a role in the recruitment and polarization of TAMs. Although more research into the immune microenvironment is warranted, there is evidence that targeting or stimulating TAMs and their factors provide a potential treatment option for DMG. In this review, we provide insight into the current status of DMG research, assess the knowledge of the immune microenvironment in DMG and GBM, and present recent findings and therapeutic opportunities surrounding the TAM–glioma crosstalk.
Collapse
Affiliation(s)
- Casper J. Pachocki
- grid.5477.10000000120346234Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M. Hol
- grid.5477.10000000120346234Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
28
|
Wei Y, Li X. Different phenotypes of microglia in animal models of Alzheimer disease. Immun Ageing 2022; 19:44. [PMID: 36209099 PMCID: PMC9547462 DOI: 10.1186/s12979-022-00300-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/19/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022]
Abstract
Microglia are immune-competent cells that are critically involved in maintaining normal brain function. A prominent characteristic of Alzheimer disease (AD) is microglial proliferation and activation concentrated around amyloid plaques in the brain. Recent research has revealed numerous microglial phenotypes related to aging and AD, apart from the traditional M1 and M2 types. Redox signalling modulates the acquisition of the classical or alternative microglia activation phenotypes. The numerous microglial functions can be achieved through these multiple phenotypes, which are associated with distinct molecular signatures.
Collapse
Affiliation(s)
- Yun Wei
- grid.464481.b0000 0004 4687 044XXiyuan Hospital of China Academy of Chinese Medical Sciences, 100091 Beijing, China
| | - Xianxiao Li
- Jingxi Cancer Hospital, 100161 Beijing, China
| |
Collapse
|
29
|
Urbantat RM, Jelgersma C, Vajkoczy P, Brandenburg S, Acker G. Combining TMZ and SB225002 induces changes of CXCR2 and VEGFR signalling in primary human endothelial cells in vitro. Oncol Rep 2022; 48:158. [PMID: 35856448 PMCID: PMC9350968 DOI: 10.3892/or.2022.8373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/27/2022] [Indexed: 11/20/2022] Open
Abstract
Standard of care therapy for glioblastoma (GBM) consisting of surgical removal, temozolomide (TMZ) and radiotherapy fails to cure the disease and median survival is limited to 15 months. Therapeutic approaches targeting vascular endothelial growth factor (VEGF)-mediated angiogenesis, one of the major drivers of tumour growth, have not prolonged patient survival as reported in clinical studies. Apart from VEGFR signalling, proangiogenic C-X-C motif chemokine receptor 2 (CXCR2) is of special interest as its ligands C-X-C motif chemokine ligand 2 (CXCL2) and interleukin-8 (IL8) are upregulated and associated with reduced survival in GBM patients. As CXCR2 is also expressed by endothelial cells, the aim of the present study was to elucidate the effect of combination therapy on gene and protein expression of primary human endothelial cells (HUVECs). To mimic the GBM specific CXCL2/IL8 oversupply environment [referred to as stimulation (STIM)], HUVECs were treated with a cocktail of CXCL2/IL8 and/or TMZ and/or CXCR2-antagonist SB225002 (SB). In brief, six treatment conditions were utilized: i) Control, ii) STIM (CXCL2/IL8), iii) TMZ + SB, iv) STIM + TMZ, v) STIM + SB, vi) STIM + TMZ + SB followed by either RNA-isolation and RT-qPCR for BAX, BCL2, vascular endothelial growth receptor (VEGFR)1/2, VEGF, CXCR1/2, CXCL2 and IL8 or immunofluorescence staining for VEGFR2 and CXCR2. SB and TMZ led to morphological changes of HUVECs and downregulated antiapoptotic BCL2 in vitro. In addition, gene expression of the alternative proangiogenic CXCL2/IL8/CXCR2 signalling pathway was significantly altered by the combination therapy, while the VEGF/VEGFR1/2 axis was only mildly affected. Furthermore, VEGFR2 and CXCR2 gene and protein expression regulation differed. VEGFR2 was not altered at the gene expression level, while combination therapy with TMZ and SB led to a 74% upregulation of VEGFR2 at the protein level. By contrast, CXCR2 was upregulated 5-fold by the combination therapy at the gene expression level and downregulated by 72.5% at the protein expression level. The present study provided first insights into the molecular changes of two major proangiogenic pathways in primary endothelial cells during treatment with TMZ and SB. Different gene and protein expression levels of the proangiogenic receptors CXCR2 and VEGFR2 in vitro must be taken into consideration in future studies.
Collapse
Affiliation(s)
- Ruth M Urbantat
- Department of Neurosurgery, Charité‑Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‑Universität zu Berlin, D‑10117 Berlin, Germany
| | - Claudius Jelgersma
- Department of Neurosurgery, Charité‑Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‑Universität zu Berlin, D‑10117 Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité‑Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‑Universität zu Berlin, D‑10117 Berlin, Germany
| | - Susan Brandenburg
- Department of Neurosurgery, Charité‑Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‑Universität zu Berlin, D‑10117 Berlin, Germany
| | - Gueliz Acker
- Department of Neurosurgery, Charité‑Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‑Universität zu Berlin, D‑10117 Berlin, Germany
| |
Collapse
|
30
|
Sansone G, Vivori N, Vivori C, Di Stefano AL, Picca A. Basic premises: searching for new targets and strategies in diffuse gliomas. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00507-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Matsuo T, Komori R, Nakatani M, Ochi S, Yokota-Nakatsuma A, Matsumoto J, Takata F, Dohgu S, Ishihara Y, Itoh K. Levetiracetam Suppresses the Infiltration of Neutrophils and Monocytes and Downregulates Many Inflammatory Cytokines during Epileptogenesis in Pilocarpine-Induced Status Epilepticus Mice. Int J Mol Sci 2022; 23:7671. [PMID: 35887020 PMCID: PMC9319101 DOI: 10.3390/ijms23147671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/30/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Acute brain inflammation after status epilepticus (SE) is involved in blood-brain barrier (BBB) dysfunction and brain edema, which cause the development of post-SE symptomatic epilepsy. Using pilocarpine-induced SE mice, we previously reported that treatment with levetiracetam (LEV) after SE suppresses increased expression levels of proinflammatory mediators during epileptogenesis and prevents the development of spontaneous recurrent seizures. However, it remains unclear how LEV suppresses neuroinflammation after SE. In this study, we demonstrated that LEV suppressed the infiltration of CD11b+CD45high cells into the brain after SE. CD11b+CD45high cells appeared in the hippocampus between 1 and 4 days after SE and contained Ly6G+Ly6C+ and Ly6G-Ly6C+ cells. Ly6G+Ly6C+ cells expressed higher levels of proinflammatory cytokines such as IL-1β and TNFα suggesting that these cells were inflammatory neutrophils. Depletion of peripheral Ly6G+Ly6C+ cells prior to SE by anti-Ly6G antibody (NIMP-R14) treatment completely suppressed the infiltration of Ly6G+Ly6C+ cells into the brain. Proteome analysis revealed the downregulation of a variety of inflammatory cytokines, which exhibited increased expression in the post-SE hippocampus. These results suggest that Ly6G+Ly6C+ neutrophils are involved in the induction of acute brain inflammation after SE. The proteome expression profile of the hippocampus treated with LEV after SE was similar to that after NIMP-R14 treatment. Therefore, LEV may prevent acute brain inflammation after SE by suppressing inflammatory neutrophil infiltration.
Collapse
Affiliation(s)
- Taira Matsuo
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Rie Komori
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Minami Nakatani
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Shiori Ochi
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Aya Yokota-Nakatsuma
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan;
| | - Junichi Matsumoto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (J.M.); (F.T.); (S.D.)
| | - Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (J.M.); (F.T.); (S.D.)
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (J.M.); (F.T.); (S.D.)
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan;
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| |
Collapse
|
32
|
Ortiz-Rivera J, Albors A, Kucheryavykh Y, Harrison JK, Kucheryavykh L. The Dynamics of Tumor-Infiltrating Myeloid Cell Activation and the Cytokine Expression Profile in a Glioma Resection Site during the Post-Surgical Period in Mice. Brain Sci 2022; 12:brainsci12070893. [PMID: 35884700 PMCID: PMC9313002 DOI: 10.3390/brainsci12070893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma is the most aggressive brain cancer and is highly infiltrated with cells of myeloid lineage (TIM) that support tumor growth and invasion. Tumor resection is the primary treatment for glioblastoma; however, the activation state of TIM at the site of tumor resection and its impact on glioma regrowth are poorly understood. Using the C57BL/6/GL261 mouse glioma implantation model, we investigated the state of TIM in the tumor resection area during the post-surgical period. TIM isolated from brain tissue at the resection site were analyzed at 0, 1, 4, 7, 14, and 21 days after tumor resection. An increase in expression of CD86 during the first 7 days after surgical resection and then upregulation of arginase 1 from the 14th to 21st days after resection were detected. Cytokine expression analysis combined with qRT-PCR revealed sustained upregulation of IL4, IL5, IL10, IL12, IL17, vascular endothelial growth factor (VEGF), and monocyte chemoattractant protein 1 (MCP1/CCL2) in TIM purified from regrown tumors compared with primary implanted tumors. Flow cytometry analysis revealed increased CD86+/CD206+ population in regrown tumors compared with primary implanted tumors. Overall, we found that TIM in primary implanted tumors and tumors regrown after resection exhibited different phenotypes and cytokine expression patterns.
Collapse
Affiliation(s)
- Jescelica Ortiz-Rivera
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
- Correspondence:
| | - Alejandro Albors
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
| | - Yuriy Kucheryavykh
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
| | - Jeffrey K. Harrison
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Lilia Kucheryavykh
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
| |
Collapse
|
33
|
Glycan-Lectin Interactions as Novel Immunosuppression Drivers in Glioblastoma. Int J Mol Sci 2022; 23:ijms23116312. [PMID: 35682991 PMCID: PMC9181495 DOI: 10.3390/ijms23116312] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/23/2022] [Accepted: 06/03/2022] [Indexed: 02/04/2023] Open
Abstract
Despite diagnostic and therapeutic improvements, glioblastoma (GB) remains one of the most threatening brain tumor in adults, underlining the urgent need of new therapeutic targets. Lectins are glycan-binding proteins that regulate several biological processes through the recognition of specific sugar motifs. Lectins and their ligands are found on immune cells, endothelial cells and, also, tumor cells, pointing out a strong correlation among immunity, tumor microenvironment and vascularization. In GB, altered glycans and lectins contribute to tumor progression and immune evasion, shaping the tumor-immune landscape promoting immunosuppressive cell subsets, such as myeloid-derived suppressor cells (MDSCs) and M2-macrophages, and affecting immunoeffector populations, such as CD8+ T cells and dendritic cells (DCs). Here, we discuss the latest knowledge on the immune cells, immune related lectin receptors (C-type lectins, Siglecs, galectins) and changes in glycosylation that are involved in immunosuppressive mechanisms in GB, highlighting their interest as possible novel therapeutical targets.
Collapse
|
34
|
Liu X, Liu Y, Qi Y, Huang Y, Hu F, Dong F, Shu K, Lei T. Signal Pathways Involved in the Interaction Between Tumor-Associated Macrophages/TAMs and Glioblastoma Cells. Front Oncol 2022; 12:822085. [PMID: 35600367 PMCID: PMC9114701 DOI: 10.3389/fonc.2022.822085] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 12/16/2022] Open
Abstract
It is commonly recognized, that glioblastoma is a large complex composed of neoplastic and non-neoplastic cells. Tumor-associated macrophages account for the majority of tumor bulk and play pivotal roles in tumor proliferation, migration, invasion, and survival. There are sophisticated interactions between malignant cells and tumor associated-macrophages. Tumor cells release a variety of chemokines, cytokines, and growth factors that subsequently lead to the recruitment of TAMs, which in return released a plethora of factors to construct an immunosuppressive and tumor-supportive microenvironment. In this article, we have reviewed the biological characteristics of glioblastoma-associated macrophages and microglia, highlighting the emerging molecular targets and related signal pathways involved in the interaction between TAMs and glioblastoma cells, as well as the potential TAMs-associated therapeutic targets for glioblastoma.
Collapse
Affiliation(s)
- Xiaojin Liu
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Liu
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiwei Qi
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yimin Huang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Hu
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangyong Dong
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Shu
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Lei
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Tumor-Associated Macrophages in Gliomas—Basic Insights and Treatment Opportunities. Cancers (Basel) 2022; 14:cancers14051319. [PMID: 35267626 PMCID: PMC8909866 DOI: 10.3390/cancers14051319] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Macrophages are a specialized immune cell type found in both invertebrates and vertebrates. Versatile in functionality, macrophages carry out important tasks such as cleaning cellular debris in healthy tissues and mounting immune responses during infection. In many cancer types, macrophages make up a significant portion of tumor tissue, and these are aptly called tumor-associated macrophages. In gliomas, a group of primary brain tumors, these macrophages are found in very high frequency. Tumor-associated macrophages can promote glioma development and influence the outcome of various therapeutic regimens. At the same time, these cells provide various potential points of intervention for therapeutic approaches in glioma patients. The significance of tumor-associated macrophages in the glioma microenvironment and potential therapeutic targets are the focus of this review. Abstract Glioma refers to a group of primary brain tumors which includes glioblastoma (GBM), astrocytoma and oligodendroglioma as major entities. Among these, GBM is the most frequent and most malignant one. The highly infiltrative nature of gliomas, and their intrinsic intra- and intertumoral heterogeneity, pose challenges towards developing effective treatments. The glioma microenvironment, in addition, is also thought to play a critical role during tumor development and treatment course. Unlike most other solid tumors, the glioma microenvironment is dominated by macrophages and microglia—collectively known as tumor-associated macrophages (TAMs). TAMs, like their homeostatic counterparts, are plastic in nature and can polarize to either pro-inflammatory or immunosuppressive states. Many lines of evidence suggest that immunosuppressive TAMs dominate the glioma microenvironment, which fosters tumor development, contributes to tumor aggressiveness and recurrence and, very importantly, impedes the therapeutic effect of various treatment regimens. However, through the development of new therapeutic strategies, TAMs can potentially be shifted towards a proinflammatory state which is of great therapeutic interest. In this review, we will discuss various aspects of TAMs in the context of glioma. The focus will be on the basic biology of TAMs in the central nervous system (CNS), potential biomarkers, critical evaluation of model systems for studying TAMs and finally, special attention will be given to the potential targeted therapeutic options that involve the TAM compartment in gliomas.
Collapse
|
36
|
Glioblastoma Microenvironment and Cellular Interactions. Cancers (Basel) 2022; 14:cancers14041092. [PMID: 35205842 PMCID: PMC8870579 DOI: 10.3390/cancers14041092] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/31/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary This paper summarizes the crosstalk between tumor/non-tumor cells and other elements of the glioblastoma (GB) microenvironment. In tumor pathology, glial cells result in the highest number of cancers, and GB is considered the most lethal tumor of the central nervous system (CNS). The tumor microenvironment (TME) is a complex peritumoral hallo composed of tumor cells and several non-tumor cells (e.g., nervous cells, stem cells, fibroblasts, vascular and immune cells), which might be a key factor for the ineffective treatment since the microenvironment modulates the biologic status of the tumor with the increase in its evasion capacity. A deeper understanding of cell–cell interactions in the TME and with the tumor cells could be the basis for a more efficient therapy. Abstract The central nervous system (CNS) represents a complex network of different cells, such as neurons, glial cells, and blood vessels. In tumor pathology, glial cells result in the highest number of cancers, and glioblastoma (GB) is considered the most lethal tumor in this region. The development of GB leads to the infiltration of healthy tissue through the interaction between all the elements of the brain network. This results in a GB microenvironment, a complex peritumoral hallo composed of tumor cells and several non-tumor cells (e.g., nervous cells, stem cells, fibroblasts, vascular and immune cells), which might be the principal factor for the ineffective treatment due to the fact that the microenvironment modulates the biologic status of the tumor with the increase in its evasion capacity. Crosstalk between glioma cells and the brain microenvironment finally inhibits the beneficial action of molecular pathways, favoring the development and invasion of the tumor and its increasing resistance to treatment. A deeper understanding of cell–cell interactions in the tumor microenvironment (TME) and with the tumor cells could be the basis for a more efficient therapy.
Collapse
|
37
|
The molecular feature of macrophages in tumor immune microenvironment of glioma patients. Comput Struct Biotechnol J 2021; 19:4603-4618. [PMID: 34471502 PMCID: PMC8383063 DOI: 10.1016/j.csbj.2021.08.019] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background Gliomas are one of the most common types of primary tumors in central nervous system. Previous studies have found that macrophages actively participate in tumor growth. Methods Weighted gene co-expression network analysis was used to identify meaningful macrophage-related gene genes for clustering. Pamr, SVM, and neural network were applied for validating clustering results. Somatic mutation and methylation were used for defining the features of identified clusters. Differentially expressed genes (DEGs) between the stratified groups after performing elastic regression and principal component analyses were used for the construction of MScores. The expression of macrophage-specific genes were evaluated in tumor microenvironment based on single cell sequencing analysis. A total of 2365 samples from 15 glioma datasets and 5842 pan-cancer samples were used for external validation of MScore. Results Macrophages were identified to be negatively associated with the survival of glioma patients. Twenty-six macrophage-specific DEGs obtained by elastic regression and PCA were highly expressed in macrophages at single-cell level. The prognostic value of MScores in glioma was validated by the active proinflammatory and metabolic profile of infiltrating microenvironment and response to immunotherapies of samples with this signature. MScores managed to stratify patient survival probabilities in 15 external glioma datasets and pan-cancer datasets, which predicted worse survival outcome. Sequencing data and immunohistochemistry of Xiangya glioma cohort confirmed the prognostic value of MScores. A prognostic model based on MScores demonstrated high accuracy rate. Conclusion Our findings strongly support a modulatory role of macrophages, especially M2 macrophages in glioma progression and warrants further experimental studies.
Collapse
Key Words
- ACC, Adrenocortical carcinoma
- BBB, brain blood barrier
- BLCA, Bladder Urothelial Carcinoma
- BRCA, Breast invasive carcinoma
- CDF, cumulative distribution function
- CESC, Cervical squamous cell carcinoma and endocervical adenocarcinoma
- CGGA, Chinese Glioma Genome Atlas
- CHOL, Cholangiocarcinoma
- CNA, copy number alternations
- CNV, copy number variation
- COAD, Colon adenocarcinoma
- CSF-1, colony-stimulating factor-1
- DLBC, Lymphoid Neoplasm Diffuse Large B-cell Lymphoma
- DMP, differentially methylated position
- ESCA, Esophageal carcinoma
- GBM, glioblastoma
- GEO, Gene Expression Omnibus
- GO, gene ontology
- GSEA, gene set enrichment analysis
- GSVA, gene set variation analysis
- Glioma microenvironment
- HNSC, Head and Neck squamous cell carcinoma
- IGR, intergenic region
- IHC, immunohistochemistry
- IL, interleukin
- Immunotherapy
- KEGG, Kyoto Encyclopaedia of Genes and Genomes
- KICH, Kidney Chromophobe
- KIRC, Kidney renal clear cell carcinoma
- KIRP, Kidney renal papillary cell carcinoma
- LGG, low grade glioma
- LIHC, Liver hepatocellular carcinoma
- LUAD, Lung adenocarcinoma
- LUSC, Lung squamous cell carcinoma
- MMP-2, matrix metalloproteinase-2
- MT1, MMP membrane type 1 matrix metalloprotease
- Machine learning
- Macrophage
- OV, Ovarian serous cystadenocarcinoma
- PAAD, Pancreatic adenocarcinoma
- PAM, partition around medoids
- PCA, principal component analysis
- PCPG, Pheochromocytoma and Paraganglioma
- PRAD, Prostate adenocarcinoma
- Prognostic model
- READ, Rectum adenocarcinoma
- SARC, Sarcoma
- SKCM, Skin Cutaneous Melanoma
- SNP, single-nucleotide polymorphism
- SNV, single-nucleotide variant
- STAD, Stomach adenocarcinoma
- SVM, Support Vector Machines
- TAM, tumor associated macrophage
- TCGA, The Cancer Genome Atlas
- TGF-β, tumor growth factor-β
- THCA, Thyroid carcinoma
- THYM, Thymoma
- TIMP-2, tissue inhibitor of metalloproteinase-2
- TLR2, toll-like receptor 2
- TME, tumor microenvironment
- TNFα, tumor necrosis factor α
- TSS, transcription start site
- UCEC, Uterine Corpus Endometrial Carcinoma
- UCS, Uterine Carcinosarcoma
- WGCNA, weighted gene co-expression network analysis
- pamr, prediction analysis for microarrays
Collapse
|
38
|
Comba A, Faisal SM, Varela ML, Hollon T, Al-Holou WN, Umemura Y, Nunez FJ, Motsch S, Castro MG, Lowenstein PR. Uncovering Spatiotemporal Heterogeneity of High-Grade Gliomas: From Disease Biology to Therapeutic Implications. Front Oncol 2021; 11:703764. [PMID: 34422657 PMCID: PMC8377724 DOI: 10.3389/fonc.2021.703764] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastomas (GBM) are the most common and aggressive tumors of the central nervous system. Rapid tumor growth and diffuse infiltration into healthy brain tissue, along with high intratumoral heterogeneity, challenge therapeutic efficacy and prognosis. A better understanding of spatiotemporal tumor heterogeneity at the histological, cellular, molecular, and dynamic levels would accelerate the development of novel treatments for this devastating brain cancer. Histologically, GBM is characterized by nuclear atypia, cellular pleomorphism, necrosis, microvascular proliferation, and pseudopalisades. At the cellular level, the glioma microenvironment comprises a heterogeneous landscape of cell populations, including tumor cells, non-transformed/reactive glial and neural cells, immune cells, mesenchymal cells, and stem cells, which support tumor growth and invasion through complex network crosstalk. Genomic and transcriptomic analyses of gliomas have revealed significant inter and intratumoral heterogeneity and insights into their molecular pathogenesis. Moreover, recent evidence suggests that diverse dynamics of collective motion patterns exist in glioma tumors, which correlate with histological features. We hypothesize that glioma heterogeneity is not stochastic, but rather arises from organized and dynamic attributes, which favor glioma malignancy and influences treatment regimens. This review highlights the importance of an integrative approach of glioma histopathological features, single-cell and spatially resolved transcriptomic and cellular dynamics to understand tumor heterogeneity and maximize therapeutic effects.
Collapse
Affiliation(s)
- Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Todd Hollon
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yoshie Umemura
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Felipe J Nunez
- Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Sebastien Motsch
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, United States
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
39
|
Qiao S, Cheng Y, Liu M, Ji Q, Zhang B, Mei Q, Liu D, Zhou S. Chemoattractants driven and microglia based biomimetic nanoparticle treating TMZ-resistant glioblastoma multiforme. J Control Release 2021; 336:54-70. [PMID: 34129862 DOI: 10.1016/j.jconrel.2021.06.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/14/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023]
Abstract
Currently, clinical treatment for temozolomide (TMZ)-resistant glioblastoma multiforme (GBM) is still a difficult problem. The aim of this paper is to set up a new GBM-targeted drug delivery system to treat TMZ-resistant GBM. Zoledronate (ZOL) not only induces apoptosis of TMZ-resistant GBM cells by down-regulation of farnesyl pyrophosphate synthetase (FPPS) but also increases the proportion of M1-type GBM associated macrophages (GAM). Based on chemoattractants secreted by GBM cells, a ZOL loaded nanoparticle coated with microglia cell membrane (ZOL@CNPs) was prepared to deliver ZOL to central nervous system to treat TMZ-resistant GBM. ZOL@CNPs was actively recruited to TMZ-resistant GBM region by CX3CL1/CX3CR1 and CSF-1/CSF-1R signal axis, and the release of ZOL from ZOL@CNPs was triggered by glutathione in GBM cells. ZOL@CNPs inhibited the growth of TMZ-resistant GBM through inducing apoptosis and inhibiting the migration and invasion of TMZ-resistant GBM cells. Besides, the immunosuppressive and hypoxic microenvironment, playing an important role in the growth of TMZ-resistant GBM, was significantly improved by ZOL@CNPs through increasing the proportion of M1-type GAM and blocking the expression of HIF-1α. ZOL@CNPs has a great potential application in the treatment for TMZ-resistant GBM.
Collapse
Affiliation(s)
- Sai Qiao
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Qibing Mei
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China.
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an 710032, China.
| |
Collapse
|
40
|
Zhang D, Hu W, Tu H, Hackfort BT, Duan B, Xiong W, Wadman MC, Li YL. Macrophage depletion in stellate ganglia alleviates cardiac sympathetic overactivation and ventricular arrhythmogenesis by attenuating neuroinflammation in heart failure. Basic Res Cardiol 2021; 116:28. [PMID: 33884509 PMCID: PMC8060235 DOI: 10.1007/s00395-021-00871-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022]
Abstract
Cardiac sympathetic overactivation is involved in arrhythmogenesis in patients with chronic heart failure (CHF). Inflammatory infiltration in the stellate ganglion (SG) is a critical factor for cardiac sympathoexcitation in patients with ventricular arrhythmias. This study aims to investigate if macrophage depletion in SGs decreases cardiac sympathetic overactivation and ventricular arrhythmogenesis in CHF. Surgical ligation of the coronary artery was used for induction of CHF. Clodronate liposomes were microinjected into bilateral SGs of CHF rats for macrophage depletion. Using cytokine array, immunofluorescence staining, and Western blot analysis, we found that macrophage expansion and expression of TNFα and IL-1β in SGs were markedly increased in CHF rats. Flow cytometry data confirmed that the percentage of macrophages in SGs was higher in CHF rats than that in sham rats. Clodronate liposomes significantly reduced CHF-elevated proinflammatory cytokine levels and macrophage expansion in SGs. Clodronate liposomes also reduced CHF-increased N-type Ca2+ currents and excitability of cardiac sympathetic postganglionic neurons and inhibited CHF-enhanced cardiac sympathetic nerve activity. ECG data from 24-h, continuous telemetry recording in conscious rats demonstrated that clodronate liposomes not only restored CHF-induced heterogeneity of ventricular electrical activities, but also decreased the incidence and duration of ventricular tachycardia/fibrillation in CHF. Macrophage depletion with clodronate liposomes attenuated CHF-induced cardiac sympathetic overactivation and ventricular arrhythmias through reduction of macrophage expansion and neuroinflammation in SGs.
Collapse
Affiliation(s)
- Dongze Zhang
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Wenfeng Hu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bryan T Hackfort
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wanfen Xiong
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Michael C Wadman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|